Advances in Radiation Oncology in Lung Cancer 2nd Edition

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 814

Medical Radiology

Radiation Oncology

Series Editors

Luther W. Brady
Hans-Peter Heilmann
Michael Molls
Carsten Nieder

For further volumes:


http://www.springer.com/series/4353
Branislav Jeremić
Editor

Advances in Radiation
Oncology in Lung Cancer
Second Edition

Foreword by
Luther W. Brady
Hans-Peter Heilmann
Michael Molls
Carsten Nieder

123
Editor
Branislav Jeremić
Institute of Lung Diseases
Institutski put 4
21204 Sremska Kamenica
Serbia
e-mail: nebareje@gmail.com

ISSN 0942-5373
ISBN 978-3-642-19924-0 e-ISBN 978-3-642-19925-7
DOI 10.1007/978-3-642-19925-7
Springer Heidelberg Dordrecht London New York

Library of Congress Control Number: 2011936026

Ó Springer-Verlag Berlin Heidelberg 2011

This work is subject to copyright. All rights are reserved, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting,
reproduction on microfilm or in any other way, and storage in data banks. Duplication of this publication
or parts thereof is permitted only under the provisions of the German Copyright Law of September 9,
1965, in its current version, and permission for use must always be obtained from Springer. Violations are
liable to prosecution under the German Copyright Law.
The use of general descriptive names, registered names, trademarks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
Product liability: The publishers cannot guarantee the accuracy of any information about dosage and
application contained in this book. In every individual case the user must check such information by
consulting the relevant literature.

Cover design: eStudio Calamar, Berlin/Figueres

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


For
Aleksandra and Marta
Foreword

Worldwide, lung cancer remains the most enigmatic and difficult malignancy to
treat in a definitive fashion. It is estimated that there are more than 1.35 million
cases in the world in 2010 representing about 13% of all malignant diseases
with which physicians have to deal. Lung cancer represents about 31% of all
cancer related deaths in the world. More people die of lung cancer than of the
next three common causes of cancer related deaths.
In spite of incredible efforts at prevention, early diagnosis, new and inno-
vative technologies relative to treatment, the outcome remains dismal with no
significant improvement in the overall five year survival rate in the last 25 years.
The survival at five years measured by surveillance, epidemiology, and end
results program is about 15%, a number which has not changed significantly
for 30 years.
The present volume edited by Professor Branislav Jeremic represents a
contemporary statement of all the aspects in the evolution of treatment of lung
cancer. It represents the opinions stated by the world’s leading lung cancer
specialists and covers not only basic science for lung cancer clinical investiga-
tions, but histology and staging, and a wide range of fundamental treatment
considerations. The effort looks at current treatment strategies for non small
cell and small cell lung cancers evaluated in the context of detail with due
attention to novel approaches that may promise further improvement in
outcome.
The various types of treatment related toxicities are discussed along with
issues of quality of life and prognostic factors.
The volume deals not only with a major statement relative to angiogenesis in
lung cancer but also the aspects of molecular biology and genetics for the
disease process, along with the clinical investigations having to do with bron-
choscopy, pathology, radiologic images, PET/CT studies for staging and
evaluation of treatment outcome as well as surgical staging.
A significant point is the role for surgery alone or in combination with
radiation therapy with or without systemically administered chemotherapy as
well as the statement relative to the impact for immunotherapy in terms of
management.
Sections deal with the basic aspects of appropriate, proper radiation therapy
administration, reviewing conventional radiotherapy along with stereotactic
radiation therapy and with data presented having to do with IMRT/IMAT,
cyberknife, tomotherapy, proton therapy, and carbon ion therapy.

vii
viii Foreword

One of the most difficult issues relative to lung cancer is the identification of
chemotherapeutic agents that would be of significant impact in changing the
outlook and prospects for survival for the patients. Data presented in the
volume deal with novel chemotherapy agents as well as novel targeted agents
and how they might best be combined to improve outcome.
The volume is an important statement of the contemporary status in the
multiple aspects of this complicated and difficult disease process and would be
an important statement for basic scientists and clinicians in all aspects of
treatment. It also makes significant statements relative to epidemiological
studies that would be appropriate in terms of efforts to establish technologies
for earlier diagnosis and more effective combined multimodal treatment
programs.

Luther W. Brady
Hans-Peter Heilmann
Michael Molls
Carsten Nieder
Preface

Lung cancer is the cancer with the highest incidence (1.61 million cases in 2008;
12.7% total cancer cases) and is the major cancer killer in both sexes across the
world (1.38 million deaths in 2008; 18.2% of total cancer mortality). Due to
moving tobacco industry focus from developed to a developing world, more
than 50% of new cases now occur in the latter one. This large shift is seen in the
past decade, with anticipated continuation of this trend as rates of cigarette
smoking continue to rise in newly industrialised countries. It is, therefore, not a
surprising fact that it represents a big burden to national health care systems
worldwide with hundreds of thousands of patients succumbing to it every year.
Radiation therapy remains the cornerstone of modern treatment approaches
nowadays. It is so in both histologies (nonsmall cell and small cell) and in all
stages of the disease. It is also so in both curative and palliative setting and is
deemed by many as the most cost-effective treatment option in lung cancer.
Being now more than 110 years old, this treatment modality passed a long way
in its technological and biological development. Integrated well with numerous
diagnostic approaches and other two important treatment options (surgery and
chemotherapy), it continuously change and adapt to the growing demands of
both modern society and successfully assimilate within the framework of
computerised domain. Indeed, there seems to be very few competitors among
medical disciplines that have so broadly embraced novel computer-driven
technological aspects as is the case with radiation therapy.
This, updated and second edition of the book initially published in 2004 is
very much about it: constant research and development in the field of radiation
oncology as the vital part (ingredient, one may say!) of our comprehensive and
very much orchestrated approach in the diagnosis and treatment of lung cancer.
While intervening seven years may deem too short for any major leaps in this
field, I am sure this second edition will stand the test of time as the necessary
checkpoint in the global development in this field.
To demonstrate this premise, the skeleton of the first edition remained,
though updated. More substance is now provided as to keep the pace with novel
technological and biological advances in the field, becoming new standards of
care almost daily. Various, non-radiation oncology aspects are also now
included in the book with the same goal. Ultimately, composition of the book is
such that it aims not only radiation oncologists but other lung cancer specialists
which, I firmly believe, would largely benefit from it.

ix
x Preface

In this effort, I have had great pleasure and very much privilege of having a
distinguished faculty joining me. The faculty that dedicated their professional
lives to the fight against lung cancer and have continuously provided substantial
contribution in this field. The faculty that have built and steamed towards the
same: more comprehensive understanding of basic premises of biology and
technology, its successful incorporation in the diagnosis and treatment of the
disease finally ending up in state-of-the-art approaches of the second decade of
the new millennium.
I would also like to thank my former and current staff colleagues with whom
I have collaborated in sometimes distant, but beautiful places. This is especially
so in cases of those still living and working in developing countries. From such
collaboration, I grew up not only as a better and more mature medical pro-
fessional, but also a better human being.
I would also like to express my gratitude to Alexander von Humboldt
Foundation, Bonn, Germany for their continuous support since 1998. Special
thanks go to Ms. Daniela Brandt and Ms. Martina Wiese from Springer who
did super job which successfully ends up here with you. Without them, the
expiring year would simply be impossible to imagine and the final shape would
not be thoroughly and timely reached.

Belgrade Branislav Jeremić


Contents

Part I Basic Science of Lung Cancer

Molecular Biology and Genetics of Lung Cancer . . . . . . . . . . . . . . . . . 3


Dusan Milanovic

Angiogenesis and Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17


Wenyin Shi and Dietmar W. Siemann

Part II Clinical Investigations

Interventional Pulmonology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Branislav Perin, Bojan Zarić, and Heinrich D. Becker

Pathology of Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53


Mary Beth Beasley

Radiologic Imaging of Lung Cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . 63


Palmi Shah and James L. Mulshine

PET/CT for Staging and Diagnosis of Lung Cancer . . . . . . . . . . . . . . . 75


Sigrid Stroobants

Surgical Staging of Lung Cancer for Advances in Radiation


Oncology of Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
Farhood Farjah and Valerie W. Rusch

Part III Basic Treatment Considerations

Lung Cancer Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103


Sidhu P. Gangadharan, Walter J. Lech, and David J. Sugarbaker

Radiation Response of the Normal Lung Tissue and Lung Tumors . . . . 119
Hiromitsu Iwata, Taro Murai, and Yuta Shibamoto

xi
xii Contents

Radiation Time, Dose, and Fractionation in the Treatment


of Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Daniel Gomez, Melenda D. Jeter, Ritsuko Komaki, and James D. Cox

3D Radiation Treatment Planning and Execution . . . . . . . . . . . . . . . . . 143


Mary K. Martel

Four-dimensional Radiation Therapy for Non-Small Cell Lung


Cancer: A Clinical Perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
Max Dahele, Johan Cuijpers, and Suresh Senan

PET and PET/CT in Treatment Planning. . . . . . . . . . . . . . . . . . . . . . . 173


Michael P. Mac Manus and Rodney J. Hicks

Target Volume Definition in Non-Small Cell Lung Cancer . . . . . . . . . . 187


Lucyna Kepka and Milena Kolodziejczyk

The Radiation Target in Small-Cell Lung Cancer. . . . . . . . . . . . . . . . . 201


Gregory M. M. Videtic

Radiation Sensitizers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213


Anthony M. Brade and Zishan Allibhai

Radioprotectors and Chemoprotectors in the Management


of Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
Ritsuko Komaki, Zhongxing Liao, James D. Cox,
Kathy A. Mason, and Luka Milas

Systemic Therapy for Lung Cancer for the Radiation Oncologist . . . . . 247
Chandra P. Belani

Combined Radiotherapy and Chemotherapy: Theoretical


Considerations and Biological Premises . . . . . . . . . . . . . . . . . . . . . . . . 267
Branislav Jeremic, Dusan Milanovic, and Nenad Filipovic

Radiotherapy and Second Generation Drugs . . . . . . . . . . . . . . . . . . . . 275


Michael Geier and Nicolaus Andratschke

Radiotherapy and Third Generation Concurrent


Chemotherapy Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
Ross Bland, Puneeth Iyengar, and Hak Choy

Part IV Current Treatment Strategies in Early-Stage Non-Small


Cell Lung Cancer

Conventional Radiation Therapy in Early Stage


Non-small-cell Lung Cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
Branislav Jeremić, Sinisa Stanic, and Slobodan Milisavljevic
Contents xiii

Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer . . . . . 343


John H. Heinzerling and Robert D. Timmerman

Postoperative Radiotherapy for Non-Small Cell Carcinoma . . . . . . . . . 363


Ellen Kim and Mitchell Machtay

PDT-Lung. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
Ron R. Allison

The Role of Radiofrequency Ablation in the Treatment


of Stage 1 Non-Small Cell Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . 381
John M. Varlotto, Julia A. Shelkey, and Rickhesvar P. Mahraj

Part V Current Treatment Strategies in Locally Advanced


Non-Small Cell Lung Cancer

Lung Dose Escalation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 399


Bradford S. Hoppe and Kenneth E. Rosenzweig

Radiochemotherapy in Locally Advanced Non-small-Cell


Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
Branislav Jeremić, Francesc Casas, and Asuncion Hervas-Moron

Tri-Modality Therapy in Locally Advanced Non-Small-Cell


Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 433
Jan P. van Meerbeeck and Elke Vandenbroucke

Prophylactic Cranial Irradiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 445


Jason Francis Lester

Palliative External Beam Thoracic Radiation Therapy


of Non-Small Cell Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453
Stein Sundstrøm

Intraoperative Radiotherapy in Lung Cancer: Methodology


(Electrons or Brachytherapy), Clinical Experiences
and Long-Term Institutional Results . . . . . . . . . . . . . . . . . . . . . . . . . . 461
Felipe A. Calvo, Javier Aristu, Sergey Usychkin, Leire Arbea,
Rosa Cañón, Ignacio Azinovic, and Rafael Martinez-Monge

Brachytherapy for Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477


A. Polo, M. Castro, A. Montero, and P. Navío

Part VI Current Treatment Strategies in Small Cell Lung Cancer

Limited-Disease Small-Cell Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . 491


Branislav Jeremić, Željko Dobrić, and Francesc Casas
xiv Contents

Radiation Therapy in Extensive Disease Small Cell Lung Cancer . . . . . 505


Branislav Jeremić and Luhua Wang

Prophylactic Cranial Irradiation in Small-Cell Lung Cancer . . . . . . . . 513


Michael C. Stauder and Yolanda I. Garces

Part VII Treatment in Specific Patient Groups and Other Settings

Radiation Therapy for Lung Cancer in Elderly . . . . . . . . . . . . . . . . . . 523


Branislav Jeremić and Željko Dobrić

Radiation Therapy for Recurrent Disease. . . . . . . . . . . . . . . . . . . . . . . 543


Branislav Jeremić, Jai Prakash Agarwal, and Sherif Abdel-Wahab

Radiation Therapy for Metastatic Disease . . . . . . . . . . . . . . . . . . . . . . 561


Dirk Rades

Advances in Supportive and Palliative Care


for Lung Cancer Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
Michael J. Simoff

Part VIII Treatment-Related Toxicity

Hematological Toxicity in Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . 597


Francesc Casas, Ferran Ferrer, and Núria Viñolas

Radiation-Induced Lung and Heart Toxicity . . . . . . . . . . . . . . . . . . . . 609


Liyi Xie, Xiaoli Yu, Zeljko Vujaskovic, Mitchell S. Anscher,
Timothy D. Shafman, Keith Miller, Robert Prosnitz, and Lawrence Marks

Spinal Cord Toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627


Timothy E. Schultheiss

Radiation Therapy-Related Toxicity: Esophagus. . . . . . . . . . . . . . . . . . 637


Voichita Bar Ad and Maria Werner-Wasik

Brain Toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 647


C. Nieder

Part IX Quality of Life Studies and Prognostic Factors

Quality of Life Outcomes in Radiotherapy of Lung Cancer . . . . . . . . . 661


M. Salim Siddiqui, Farzan Siddiqui, and Benjamin Movsas

Prognostic Factors in Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . 675


Frank B. Zimmermann
Contents xv

Part X Technological Advances in Lung Cancer

Intensity-Modulated Radiation Therapy and Volumetric-Modulated


Arc Therapy for Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 691
Inga S. Grills and Victor S. Mangona

Image-Guided Robotic Stereotactic Ablative Radiotherapy


for Lung Tumors: The CyberKnife . . . . . . . . . . . . . . . . . . . . . . . . . . . 715
Billy W. Loo Jr. and Iris C. Gibbs

Advances in Radiation Oncology of Lung Cancer . . . . . . . . . . . . . . . . . 725


Deepak Khuntia and Minesh P. Mehta

Image-Guided Radiotherapy in Lung Cancer . . . . . . . . . . . . . . . . . . . . 735


Percy Lee and Patrick Kupelian

Proton Therapy for Lung Cancer: State of the Science . . . . . . . . . . . . . 743


David A. Bush

Carbon Ion Radiotherapy in Hypo-Fractionation Regimen


and Single Dose for Stage I Non-small-Cell Lung Cancer . . . . . . . . . . . 753
T. Miyamoto, N. Yamamoto, M. Baba, and T. Kamada

Part XI Biological Advances in Lung Cancer

Novel Cytotoxic Agents in Combination with Radiation


in the Management of Locally Advanced Non-Small
Cell Lung Cancer: Focus on Pemetrexed
and Nab-Paclitaxel [Abraxane] . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
Corey J. Langer

Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer. . . . 773


Martin J. Edelman and Nadia Ijaz

Part XII Clinical Research in Lung Cancer

Translational Research in Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . 793


Deepinder Singh, Kevin Bylund, and Yuhchyau Chen

Clinical Research in Radiation Oncology of Lung Cancer:


Why We Fail(ed)? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809
Branislav Jeremić

Pitfalls in the Design, Analysis, Presentation, and Interpretation


of Randomized Clinical Trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 819
Richard Stephens
xvi Contents

New Directions in the Evaluation and Presentation


of Clinical Research in Lung Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . 827
Noelle O’Rourke, Fergus Macbeth, and Elinor Thompson

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837
Contributors

Ron R. Allison Greenville, NC, USA; e-mail: ALLISONR@ecu.edu


Nicolaus Andraschke Munich, Germany; e-mail: n_andratschke@yahoo.de
Mary Beth Beasley New York, NY, USA; e-mail: mbbeasleymd@yahoo.com
Chandra P. Belani Hershey, PA, USA; e-mail: cbelani@hmc.psu.edu
William Blackstock Winston Salem, NC, USA; e-mail: ablackst@wfubmc.edu
Anthony Brade Toronto, Canada; e-mail: Anthony.brade@rmp.uhn.on.ca
David Bush Loma Linda, CA, USA; e-mail: dbush@dominion.llumc.edu
Felipe Calvo Madrid, Spain; e-mail: fcalvo.hgugm@salud.madrid.org
Francesc Casas Barcelona, Spain; e-mail: fcasas@clinic.ub.es
Y. Chen e-mail: yuhchyau_chen@urmc.rochester.edu
Hak Choy Dallas, TX, USA; e-mail: Hak.Choy@utsouthwestern.edu
James D. Cox Houston, TX, USA; e-mail: jcox@mdanderson.org
Martin Edelman Baltimore, MD, USA; e-mail: medelman@umm.edu
Yolande Garces Rochester, MN, USA; e-mail: garces.yolanda@mayo.edu
Inga Grills Royal Oak, MI, USA; e-mail: igrills@beaumont.edu
Lucyna Kepka Warsaw, Poland; e-mail: lucynak@coi.waw.pl
Ritsuko Komaki Houston, TX, USA; e-mail: rkomaki@mdanderson.org
Patrick Kupelian Los Angeles, LA, USA; e-mail: pkupelian@mednet.ucla.edu
Corey Langer Philadelphia, PA, USA; e-mail: corey.langer@uphs.upenn.edu
Percy Lee Los Angeles, LA, USA; e-mail: percylee@mednet.ucla.edu
Jason Lester Cardiff, Wales, UK; e-mail: jason.lester@velindre-tr.wales.nhs.uk
Bill Loo Stanford, CA, USA; e-mail: bwloo@stanford.edu
Fergus R. Macbeth Cardiff, Wales, UK; e-mail: Fergus.Macbeth@nice.org.uk
Mitchell Machtay Cleveland, OH, USA; e-mail: mitchell.machtay@
uhhospitals.org

xvii
xviii Contributors

M. McManus e-mail: michael.macmanus@petermac.org


Lawrence B. Marks Chapel Hill, NC, USA; e-mail: marks@med.unc.edu
Mary K. Martel Houston, TX, USA; e-mail: mmartel@mdanderson.org
Minesh P. Mehta Chicago, IL, USA; e-mail: mineshpmehta@gmail.com
Tadaaki Miyamoto Chiba, Japan; e-mail: t-miyamoto@min-iren-c.or.jp
Benjamin Movsas Detroit, MI, USA; e-mail: BMOVSAS1@hfhs.org
James L. Mulshine Chicago, IL, USA; e-mail: James_L_Mulshine@rush.edu
Carsten Nieder Bodo, Norway; e-mail: Carsten.Nieder@nordlandssykehuset.no
Branislav Perin Sremska Kamenica, Serbia; e-mail: bperin@eunet.rs
Alfredo Pol Madrid, Spain, e-mail: alfredo.polo@mac.com
Dirk Rades Luebeck, Germany; e-mail: Rades.Dirk@gmx.net
Kenneth Rosenzweig New York, NY, USA; e-mail: ken.rosenzweig@
mountsinai.org
Valerie Rusch New York, NY, USA; e-mail: ruschv@mskcc.org
Dietmar W. Siemann Gainsville, FL, USA; e-mail: siemadw@ufl.edu
Michael Simoff Detroit, MI, USA; e-mail: msimoff1@hfhs.org
Richard Stephens London, UK; e-mail: richardjamesstephens@gmail.com
Timothy E. Schultheiss Duarte, CA, USA; e-mail: schultheiss@coh.org
Suresh Senan Amsterdam, The Netherlands; e-mail: s.senan@vumc.nl
Yuta Shibamoto Nagoya , Japan; e-mail: yshiba@med.nagoya-cu.ac.jp
Stein Sundstrom Trondheim, Norway; e-mail: Stein.Sundstrom@stolav.no
Sigrid Stroobants Antwerp, Belgium; e-mail: Sigrid.Stroobants@ua.ac.be
David J. Sugarbaker Boston, MA, USA; e-mail: dsugarbaker@partners.org
Robert Timmerman Dallas, TX, USA; e-mail: robert.timmerman@
utsouthwestern.edu
Jan Van Meerbeeck Ghent, Belgium; e-mail: Jan.VanMeerbeeck@uzgent.be
John Varlotto Hershey, PA, USA; e-mail: jvarlotto@hmc.psu.edu
Gregory Videtic Cleveland, OH, USA; e-mail: VIDETIG@ccf.org
Maria Werner-Wasik Philadelphia, PA, USA; e-mail: maria.werner-wasik@
mail.tju.edu
Frank B. Zimmermann Basel, Switzerland; e-mail: FZimmermann@uhbs.ch
Molecular Biology and Genetics
of Lung Cancer
Dusan Milanovic

Contents 12 Other Factors ........................................................... 12


13 Conclusion ................................................................ 13
1 Introduction.............................................................. 3 References.......................................................................... 13
2 Genetical Changes Detected with Microarray..... 4
3 Nkx2.1 ....................................................................... 5
Abstract
4 EML4-ALK .............................................................. 6 DNA Microarray technology allowed analysis of
5 LKB 1........................................................................ 7 gene expression profile of lung cancer (NSCLC
and SCLC). Better understanding of the molecu-
6 c-MYC ....................................................................... 7
lar and biological basis of this disease has led to
7 KRAS ........................................................................ 8 the identification of a number of druggable
8 Bcl-2........................................................................... 9 targets. In the last years, targeted therapies with
9 MET .......................................................................... 10 small molecule kinase inhibitors showed promis-
ing clinical activity in lung carcinoma but after
10 EGFR ........................................................................ 11
some period of time development of resistance
11 p53 ............................................................................. 12 was common event observed in patients treated
with these drugs. There is an urgent need not only
to clarify important molecular-biological mecha-
nism that contributes to the development of
resistance to molecular targeted therapies but also
to identify other important druggable targets
which are crucial for development and progres-
sion of this disease.

1 Introduction

Lung carcinoma is a disease which is characterized


with several genetical and molecular biological
changes which contributed to carcinogenesis by the
D. Milanovic (&) activation of oncogenes or inactivation of tumor
Department of Radiation Oncology,
suppressor genes. In this chapter mainly genetical and
University Hospital Freiburg,
Robert Koch Strasse 3, 79106 Freiburg, Germany molecular biological changes which are detected in
e-mail: dusan.milanovic@uniklinik-freiburg.de past 6–7 years will be discussed.

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 3
DOI: 10.1007/174_2011_310, Ó Springer-Verlag Berlin Heidelberg 2011
4 D. Milanovic

degrading cyclic AMP in airway epithelial cells) sin-


2 Genetical Changes Detected gle-gene deletion occurs within AUTS2, a gene of
with Microarray unknown function in chromosome 7q11.22. Somatic
mutations in AUTS2 or PDE4D were not detected
To detect genetic changes in lung carcinoma single while in 11 of 188 samples of PTRD somatic mutations
nucleotide polymorphism (SNP) arrays was per- were identified—three of the mutations encode pre-
formed and 51 NSCLC primary tumor samples, 26 dicted inactivating changes in the tyrosine phosphatase
NSCLC cell line samples, 19 SCLC primary tumor domain indicating that PTPRD is one probable cancer-
samples and 5 SCLC cell line samples, were analyzed associated gene. The TP53 locus which is normaly
(Zhao et al. 2005). In both NSCLC and SCLC, the mutated in *50% of lung adenocarcinomas in this data
most frequent copy number gains (C3 copies) were series shows no homozygous deletions. The amplifi-
found in chromosome arm 5p (NSCLC 25%, SCLC cation events were observed in 1–7% of all samples.
43%) while maximum degrees of copy number loss at Fourteen of the 24 regions of recurrent amplification
a given locus chromosome where detected most fre- contained a known protooncogene but only EGFR,
quently in chromosome arms 8p (33%) and 9p (26%) KRAS and ERB2 was earlier reported to be mutated in
in NSCLC and in chromosome arms 3p (68%) and 4q lung adenocarcinoma. The amplification peak was
(58%) in SCLC. In one SCLC cell-line quantitative detected on chromosome 5p to the telomerase catalytic
realtime PCR revealed of the amplification of subunit gene, TERT.
8q12–13 region (copy number of 89.9). In two SCLC Eight tumors with amplicons (a piece of DNA
primary tumor samples, this amplification was also formed as the product of natural or artificial amplifi-
found. 12p11 amplification was detected in two cation events) in chromosome 5p15 delineate a region
NSCLC specimens and 22q11 amplification in four containing ten genes including TERT. These results
NSCLC specimens. High level amplifications of indicate that TERT may be the target of the amplifi-
tyrosin kinase genes were found in three NSCLC cation contributing to cellular immortalization.
tumor specimens for EGFR (independent of kinase Chromosome 6p21.1 was focal amplificated in four
domain mutational status), in two specimens for samples in a region containing two genes, one of
FGFR1 and in one specimen for ERBB2 and MET. which (VEGFA) encodes vascular endothelial growth
In one other microarray analysis from samples of factor suggesting a possible mechanism for increased
21 patients with lung squamous cell cancer, HSN, angiogenesis and response to antiangiogenic therapy.
GCS, BTAK, TTK, cyclin E2 and NLK gene were Amplification of regions containing several cell cycle
highly upregulated and while ANG, PTPTM, and genes, such as CDK4, CDK6 and CCND1was also
C1-Inh were highly down-regulated (Talbot et al. observed.
2005). These findings were validated with reverse Amplification of chromosome 14q13.3, which
transcription-PCR. contain only MBIP and NKX2-1 genes was found in
Other groups were analyzed 371 samples of lung *12% of samples. Based on genomic and functional
adenocarcinoma (ADC) using SNP arrays (Weir et al. analyzes, NKX2-1 (NK2 homeobox 1 TITF1),was
2007). They detected a total of 57 significantly recur- identified as a novel proto-oncogene candidate which
rent events. The most frequent genomic alteration was found in ca.12% samples of lung adenocarci-
in lung adenocarcinoma was a copy-number gain of noma. This gene is located in the minimal 14q13.3
chromosome 5p, which is found in 60% of total sam- amplification interval and encodes a lineage-specific
ples. The most significant focal deletions, detected in transcription factor.
3% of all samples, involve CDKN2A/CDKN2B, well- Using array comparative genomic hybridization,
known tumor suppressor genes localized on chromo- karyotype analysis of 33 small-cell lung cancer
some 9p21 which protein products inhibit Cdk4 and (SCLC) tumors, 13 SCLC cell lines, 19 bronchial
Cdk6 cyclin dependet kinases. Three additional dele- carcinoids was performed (Voortman et al. 2010).
tions were also detected—deletions of the 50 untrans- In SCLC tumors, recurrent copy number (CN) gains
lated region of PTPRD (a gene encoding a tyrosine were observed on chromosomes 1, 3q, 5p, 6p, 12, 14,
phosphatase), homozygous deletions of PDE4D 17q, 18, 19 and 20 and recurrent CN losses on
(encodes the major phosphodiesterase responsible for chromosomes 3p, 4, 5q, 10, 13, 16q and 17p.
Molecular Biology and Genetics of Lung Cancer 5

In bronchial carcinoids, recurrent CN gains were embryogenesis, may become an oncogene when it is
detected on chromosomes 5, 7 and 14, and recurrent deregulated in certain genetic contexts. It has been
CN losses were observed on chromosomes 3, 11 and shown that a population of lung ADC cell lines
22q. In SCLC tumors, genes encoding members of the expressing Nkx2.1, which most probably represent
PI3K-AKT-mTOR pathway and apoptotic regulating cells originating from the TRU lineage, showed clear
proteins had relatively high frequencies of gene copy dependence on the persistent Nkx2.1 expression
number alterations (CNA). (Tanaka et al. 2007). The specific and significant
growth inhibition and apoptosis in theses cells were
induced when Nkx2.1 was inhibited by RNA inter-
3 Nkx2.1 ference (RNAi). On the other hand, in the same study
it has been reported that in cohort consisting of 214
Nkx2-1 also known as the thyroid transcription factor patients with NSCLC, including 174 adenocams only
1 (TTF-1) or thyroid-specific enhancer-binding pro- in four (2.3%) patients an increase amount of more
tein is one NK2-related homeobox transcription fac- than 2.5-fold of the Nkx2.1 gene was detected. One
tor. This is a 38-kDa nuclear protein that is encoded tendency of higher frequency of increased gene cop-
by a gene located on chromosome 14q13 (Guazzi ies at metastatic sites than at primary sites has been
et al. 1990). During early embryogenesis is this observed.
expressed in the thyroid, lung bronchial epithelium, The aim of one meta-analysis was to assess the
ventral neuroepithelium and ganglionic eminence of possible role of Nkx2.1 as prognostic factor for sur-
the forebrain. Mice embryo formed lobar bronchy by vival in subgroup of patients with lung ADC
embryonic day 11.5–12 which start branching to form (Berghmans et al. 2006). Eight studies were meta-
segmental bronchy by embryonic day 12.5–13. In analyzed and it has been observed that Nkx2.1 posi-
Nkx2.1 homozygous mice branching of lobar bronchy tivity is associated with better survival in NSCLC,
did not happen. This failure is still evident in mainly in early- and locally-advanced stages and in
embryonic day 14–15 (Kimura et al. 1996). It has adenocarcinomas.
been found that Nkx2.1 is expressed consistently In one lung adenocarcinoma model, which was
throughout the life stages and uniformly in the ter- induced by lentiviral-mediated somatic activation of
minal respiratory unit (TUR), which contain periph- oncogenic Kras and deletion of p53, each mouse
eral airway cells and small-sized bronchioles (Yatabe developed between 5 and 20 lung tumors (Winslow
et al. 2002). In one clinical study, 72% of investigated et al. 2011). Despite the synchronously initiations of
64 specimen of lung ADC expressed Nkx2.1 that tumors, only a subset of them becomes malignant
showed high correlation with surfactant apoprotein suggesting that malignant progression requires addi-
(Yatabe et al. 2002). Morphologically, these parts tional changes on molecular level. Mice lived
with Nkx2.1 expression were similar to terminal 8–14 months after tumor initiation and developed
TUR. The TTF-1-positive and -negative ADC based macroscopic metastases to the draining lymph nodes,
on their clinicopathologic features and expression of pleura, kidneys, heart, adrenal glands and liver. With
various cancer-associated genes showed important identification of the lentiviral integration sites was
difference—TF-1-positive ADC were prevalent of possible to a make difference between metastatic and
female and nonsmoker. Also negative p53 staining, non-metastatic tumors and to determine gene
less frequent RB loss and preserved expression of p27 expression alterations. Cross-species analysis identi-
were observed. The authors conclude that Nkx2.1 fied Nkx2.1 as a candidate suppressor of malignant
may be a lineage marker for TUR and suggested that progression. In this mice model, low Nkx2.1 expres-
molecular pathogenesis may be partially character- sion was characterized with high malignant and
ized by cellular lineage. Nkx2.1 may be a candidate poorly-differentiated primary tumors. In non-meta-
for ‘‘lineage-survival oncogene’’, which could explain static primary tumors, Nkx2.1 expression was a
‘‘lineage-specific dependency’’ mechanism in carci- 10-fold higher in metastic. In almost all lymph nodes
nogenesis (Garraway and Sellers 2006). It means that and distant macrometastases Nkx2-1 expression was
some specific transcription factor, which is a master low/absent. Expression of exogenous Nkx2-1 in
regulator of the specific cellular lineage during tumors, which were induced with lentiviral vector
6 D. Milanovic

expressing Nkx2-1, restricted tumor progression, The coiled-coil domain within this portion of EML4
resulting in fewer tumors of advanced-histopatholo- is responsible for constitutive dimerization and acti-
gical grades. In gain- and loss-of-function experi- vation of EML4-ALK.
ments in cells derived from metastatic to non- Overexpression of EML4-ALK in mouse 3T3
metastatic tumors have demonstrated that Nkx2-1 fibroblasts caused development of transformed foci in
controls tumor differentiation and limits metastatic cell culture and subcutaneous tumors in nude mice
potential in vivo. With different functional analysis (Soda et al. 2007). To investigate role of EML4-ALK
was shown that Nkx2-1 inhibits tumor growth partly in lung carcinogenesis one transgenic mice model was
by repressing the embryonically restricted chromatin established (Soda et al. 2008). Remarkably, all of
regulator Hmga2, which is highly expressed in investigated mice, which expressed EML4-ALK
embryonic lung but not in any normal adult lung cells. specifically in lung alveolar epithelial cells, devel-
On the other hand, they are a lot of functional oped hundreds of adenocarcinoma nodules in both
clinical data supporting oncogenic activity of NKX2.1 lungs within a few weeks after birth. The treatment of
in lung ADC where it is focally ampflied in *10% of mice with oral specific small molecule inhibitor of the
samples (Kwei et al. 2008). In conclusion, NKX2.1 kinase activity of ALK resulted in rapid disappear-
can have both oncogenic and tumor suppressive ance of tumors. These results were confirmed as
functions in lung ADC, demonstrate its role as a dual important carcinogenic role of EML4–ALK.
function lineage factor. In humans, EML4–ALK fusion gene is not observed
The implications, which could have NKX2.1 in frequently; it may be detected only in 2 to 7% of all
treatment of lung ADC, warrant further preclinical NSCLC patients. The EML4-ALK translocation is most
studies. Normally in adults, expression of NKX2.1 is frequently seen in patients with adenocarcinomas, in
localized in peripheral airway cells and small-sized young adults, and in patients who have never smoked
bronchioles and it is questionable, if this target is or who are light smokers (Koivunen et al. 2008).
druggable and how any pharmacolgogical manipula- Interestingly, this translocation is generally detected in
tion with NKX2.1 will influence lung function. NSCLC with wild type RAS or EGFR (Wong et al.
2009). The treatment of NSCLC patients carrying the
ALK fusion gene with crizotinib, an high specific,
4 EML4-ALK small molecule ALK inhibitor, caused tumors shrink-
age or stabilization in 90% of 82 patients (Kwak
The echinoderm microtubule-associated protein-like et al. 2010).
4 (EML4)-anaplastic lymphoma kinase (ALK) fusion Despite the early impressive response to this
oncogene (EML4-ALK) is an aberrant fusion gene therapy, development of resistance was observed
which represents one of the newest target oncogen in usually within 1 year (Choi et al. 2010). In in vitro
NSCLC. It encodes a cytoplasmic chimeric protein condition, one model was established in which
with constitutive kinase activity. EML4 and ALK resistance in highly sensitive EML4-ALK–positive
are localized oppositely oriented on short arm of NSCLC cell was generated by increasing doses of
chromosome 2 (2p21 and 2p23); either gene may be crizotinib (Katayama et al. 2011). Cells which were
inverted to generate their common fusion gene resistant to intermediate doses of crizotinib developed
product. amplification of the EML4-ALK gene while develop-
The derived protein consists of the amino-terminal ment of a gatekeeper mutation, L1196M, within the
portion of EML4 and the intracellular region of the kinase domain, was the main reason for resistance to
ALK. EML 4 consist of an N-terminal basic region, a higher crizotinib doses. The same mutation was
hydrophobic domain (HELP) for the association to described by one patient with EML4-ALK-positive
microtubules and the WD repeats that is necessary for non-small-cell lung cancer who became resistant to
protein-interactions. crizotinib after successful treatment for 5 months.
In EML4-ALK fusion protein, the N-terminal half Treatment with crizotinib was ineffective against cells
of EML4 including basic region, the HELP domain harboring this mutation (Choi et al. 2010). The
and portion of the WD-repeat region, is fused to treatment of these cells or human NSCLC H3122 CR
the intracellular juxtamembrane region of ALK. Xenografts, which were induced by implantation of
Molecular Biology and Genetics of Lung Cancer 7

crizotinib pretreated and resistant cells, with other metastasis and lived shorter than mice which have p53
two structurally different ALK inhibitors, NVP- or Ink4a/Arf deficient tumors. In case of LKB1
TAE684 and AP26113, were highly active. Interest- reconstitution in NSCLC lines lacking p16INK4a, ARF
ingly, these resistant cells harboring gatekeeper and p53 antitumors effects were observed, suggesting
mutation, L1196M were highly sensitive to the Hsp90 that LKB1 anti-tumor effects are p16INK4a, ARF and
inhibitor 17-AAG (Katayama et al. 2011). p53 independent. The anti-metastatic effect of
Despite the development of resistance to the first LKB1was also INK4a/ARF and p53 independent. In
class of EML4-ALK inhibitors this fusion product the same study, 144 human NSCLC samples were
remains a very attractive target for future drug analyzed for KRAS and LKB1 mutations; 27 of 80% of
development. Based on preclinical findings, combi- ADCs (34%) harbored predominantly (19/80) single-
nation of EML4-ALK inhibitors with Hsp90 inhibi- copy mutation or deletion. The same alteration of
tors, which are already investigated in Phase I–II LKB1 was also observed in other histological types of
clinical trials, could represent an attractive strategy to NSCLC—in 8 of 42 samples of squamous cell carci-
overcome the resistance. noma, 6 showed single-copy mutation or deletion as a
predominant lesion. NEDD9, VEGFC and CD24 were
identified as targets of LKB1 repression in human lung
5 LKB 1 cancer cell lines and mouse lung tumors. In other study
it was found that LKB1 genetic alterations favorably
LKB1 also known as Serine/threonine kinase 11 (STK appear in a subset of poorly-differentiated lung ADC
11) is a protein kinase which plays essential role from smoking male patients (Matsumoto et al. 2007).
in the regulation of the cell energetic checkpoint To explain potential role in treatment of lung
through the phosphorylation and activation of aden- carcinoma, additionally in vitro and in vivo experi-
osine monophosphate-dependent kinase (AMPK). In ments are necessary.
humans, LKB1 is encoded by LKB1/STK 11 gene,
which is localized on 19p chromosome (Jansen et al.
2009). Germline mutation of LKB1/STK11 gene is 6 c-MYC
responsible for appearance of Peutz–Jeghers or
hereditary intestinal polyposis syndrome (Beggs et al. c-MYC proto-oncogene is located on chromosome
2010). This autosomal dominant disorder is charac- 8 in humans and encodes an MYC transcription
terized by development of benign hamartomatous factor (Vennstrom et al. 1982). It is assumed that
polyps in the gastrointestinal tract and mucocutaneous c-MYC regulate expression of 15% of all genes.
pigmented lesions occurring on different locations of During the emrbyogenesis c-MYC is broadlly
the body, most frequently around/in mouth. Also expressed in different tissues and targeted gene dis-
persons with a this syndrome have an increased risk ruption of both c-MYC alleles in embryonic stem
for developing colorectal, stomach, pancreas, breast, cells caused embryonic lethality at day 9.5–10.5
lungs, ovaries, uterus and testicles carcinoma. indicating crucial MYC roll in development (Davis
LKB1 inactivation are common event in lung ADC. et al. 1993). In adults, MYC is expressed in tissues
It has been shown that 33% (6 of 20 primary tumors and that posses high proliferative capacity. In normal
two of four cell lines) of lung ADC harbor LKB1/STK11 dividing cells, c-MYC expression is maintained at a
gene inactivation. By mapping of the short arm of relatively constant intermediate level. In neoplastic
chromosome 19 from lung ADC samples it has been disease, where MYC is activated, its expression is
found that LKB1/STK11 gene is located in the minimal- moderate to very high (Pelengaris et al. 2002) .
deleted region (Sanchez-Cespedes et al. 2002). The MYC family of transcription factors contain the
tumor-suppressor function of Lkb1 was investigated in carboxy-terminal basic-helix-loop-helix-zipper
one mouse model (Ji et al. 2007). Previously it has been (bHLHZ) domain. bHLH domain binds to DNA
presumed that the tumor-suppressor activity of LKB1 is while zipper domain allows the dimerization with
caused by activation of p53 and/or the Ink4a/Arf locus. its partner Max, another bHLH transcription factor
In this model, mice with tumors, characterized with (Amati et al. 1993). This dimerization is necessary for
LKB1 loss and Kras activation, developed more Myc-dependent transactivation and its oncogenic and
8 D. Milanovic

Fig. 1 Signaling pathways


involved in the pathogenesis DNA
Activation/Production
Inhibition
of lung carcinoma Damage Mechanism unknown
HDM2
p21(ARF)
p14(ARF) p16(INK4A)
Cell
Cycle p53 Cdk4/6:cyclin D
RTK IGF
Telomerase E2F RB:E2F PTEN

Akt PI3K
Gene
Expression MAPK MEK Raf Ras RTK Growth Factors

Rho Rac
Myc:Mad
Receptor Neuropeptides
Myc:Max
Cells CAM Bax
Apoptosis Bcl-2
ECM Integrins

proliferative properties. Additionally to its role as a that this work for the first time provides information
transcriptional factor, MYC also has a fundamental of MYC role in the maintenance of Ras-dependent
role global chromatin structure by regulating histone lung tumors in vivo. The side effects of systemic
acetylation both in gene-rich regions and at sites MYC inhibition were observed in normal regenerat-
far from any known gene. On the other hand, ing tissues, but they were well rapidly and completely
MYC overexpression can activate the proapoptotic reversible (Soucek et al. 2008). In one study, genetic
BCL2 family protein BAX, resulting in release of alterations that define prognosis of patients with
cytochrome c and apoptosis (Pelengaris et al. 2002). early-stage lung ADC were investigated (Iwakawa
To attenuate the antitumorigenic effects of MYC et al. 2011). Hundred and sixty-two speciemens of
other compensatory genetic and epigenetic alterations stage I lung ADC were analyzed. It has been found
are required to facilitate tumor growth. It has been found that MYC amplification correlated with poor prog-
that overexpression of an antiapoptotic protein MCL 1, nosis. These authors conclude that MYC amplifica-
stimulates tumor progression in transgenic mice with tion may be used as a prognostic marker of patients
either spontaneous mutations in Kras or experimental with early-stage lung ADC (Fig. 1).
introduction of activated RAS only in case of MYC To determine possible role of MYC inhibition as a
paralell overexpression (Allen et al. 2011). By patients novel therapeutic strategy, additional in vivo experi-
with NSCLC, MCL1 overexpression alone was not a ments should be performed specially concering its
prognostic marker, but if overexpression of MCL 1 was safety profile.
accompanied by MYC overexpression, significantly
poorer overall survival was observed.
Principally, Myc could be an attractive therapeutic 7 KRAS
target for treating a different type of neoplastic dis-
ease, but on the other hand it would be possible that The proto-oncogene KRAS, together with HRAS
therapeutic inhibition of myc may inhibit proliferation and NRAS belongs to the Ras subfamily of small
of normal cells resulting in serious side effects. GTP-ase. RAS plays a very important role in the
Abnormal expression is observed in *20% of EGFR downstream signaling cascade and can activate
NSCLC and *30% of SCLC (Salgia and Skarin 1998). other important pathways such the serine/threonine
Systemic MYC inhibition by reversible, systemic kinase RAF, mitogen-activated kinases ERK1 and
expressed of a dominant-interfering MYC mutant was ERK2, phosphatidyl inositol 3-kinase and other pro-
investigated in one Ras-induced lung ADC mouse teins that translocate to the nucleus resulting in the
model. As a result, incipient and established lung transcription and cell proliferation (Vakiani and Solit
tumors were rapidly regressed. It is very interesting 2011).
Molecular Biology and Genetics of Lung Cancer 9

RAS can be activated by mutations in different Table 1 Molecular abnormalities in lung cancer
tumor types. Approximately 20% of human tumors Frequency of Abnormality (%)a
have activating point mutations in RAS (usually in NSCLC SCLC
codons 12, 13 and 61). About 85% of all RAS
Growth Signals
mutations are found in KRAS, 15% in NRAS and
Ras 25 \1
less than 1% in HRAS (Downward 2003). KRAS
mutation is detected in 10–30% of lung adeno- Akt 70–90 65
carcinomas, is very rarely found in other NSCLC Myc 20–60 20–30
form and is not present in SCLC. It has been EGFR 50 0–50
found that TBK1, non-canonical IjB kinase, HER2/neu 30 30
which activate antiapoptotic signals involving c-Kit 30–40 50
cREL and BCL-XL is essential for development of Neuropeptides *50 *50
KRAS-driven lung tumors (Barbie et al. 2009). IGF *90 *90
KRAS can induce IL-8 overexpression, one che-
Tumor Suppressor Genes
mokine which plays important role in cell growth,
RB 15–30 [90
tumor cells migration and angiogenesis (Sunaga
et al. 2011). In 89 NSCLC surgical specimens IL-8 p16(INK4A) inactivation 50–70 0–20
expression was significantly higher in male 3p deletions 70 90
patients, smokers, elderly patients, in patients with FHIT inactivation 40–70 70
pleural involvement and KRAS mutated adeno- RASSF1A silencing 50 90
carcinomas. In one study, the relationship between Apoptosis
KRAS amplification, detected by Fluorescence in p53 40–50 60–75
situ hybridization and mutation was investigated in
Bcl-2 20–35 71
two cohorts of patients. In one cohort (538 Swiss
Replicative Potential
patients) the prevalence of KRAS amplification
was 13.7%; in other cohort (402 patients from Telomerase 80–100 80–100
New York) the prevalence of KRAS amplification Angiogenesis
was 15.1%. Patient with KRAS amplification had a VEGF 75 75
larger, less-differentiated and aggressive tumors, COX-2 [70 Not reported
with characteristic angiolymphatic invasion. Metastasis
Especially in adenocarcinoma subset harboring N-CAM, non-adhesive not reported 90
activating KRAS mutations, KRAS amplification
Laminin-5 inactivation 20–60 65–85
was observed, suggesting a synergistic relationship a
See text for selected references
between amplification and mutation. The presence
of KRAS amplification was not associated with
nodal involvement or survival. The prognostic role
of KRAS mutations in NSCLC have been inves-
tigated in various clinical studies. Despite of 8 Bcl-2
conflicting results, a meta-analysis has revealed
that RAS mutations may have some prognostic Bcl-2 is a member of Bcl-2 genes family which plays
value for determination of a poor prognosis an important role in the regulation of apoptosis. Bcl-2
(Mascaux et al. 2005). On the other hand, one is encoded by BCL-2 gene which has been implicated
review data strongly suggests that KRAS muta- in a different neoplastic disease including lung
tions cannot be used as a factor which can predict carcinoma. So far, 15 mammalian family members
response to conventional chemotherapy (Loriot divided in three subfamilies were identified—Bcl-2,
et al. 2009). Because of its interaction with dif- BAX and BH 3. Some of them act as proapoptotic
ferent crucial pathways involved in carcinogenesis, (BAX and BH3 family members) some as antiapo-
KRAS inhibition could represent an attractive ptotic factors (Bcl-2 family members). The product of
therapeutic approach Table 1. bcl-2 is a 26 kDa protein which is mainly located in
10 D. Milanovic

the inner mitochondrial membrane. The most impor- Tyr 1234 and Tyr 1235, which involve different signal
tant function of Bcl-2 is an inhibition of apoptosis and transducers such as GRB2, SHC, SRC and the p85
prolongation of cell survival by arresting cells in the regulatory subunit of phosphatidylinositol-3 kinase
G0/G1 phase of cell cycle. The ratio of proapoptotic (PI3K) and Gab1. Gab1, member of protein-docking
to antiapoptotic members will determinate the cells family, is a most important coordinator of cellular
response for apoptotic signal (Chao and Korsmeyer response to MET. The interaction between Gab1 and
1998). Bcl-2 overexpression is frequently observed in MET causes Gab1 phosphorylation which is respon-
SCLC (*90%) (Jiang et al. 1995), where it can be sible for activation of multiple signal transduction
found seldom in patients with NSCLC (*25%) pathway such as RAS, PI3K, STAT, beta-catenin and
(Pezzella et al. 1993). It has been demonstrated in in Notch (Birchmeier et al. 2003).
vitro conditon that in SCLC cell lines which were HGF/SF1 plays a fundamental role in embryolog-
resistant to conventional chemotherapeutics Bcl-2 ical development; mice lacking HGF/SF1 cannot
was upregulated. In phase I dose-escalation trial complete development and die in utero. The most
in patients with heavily pretreated SCLC, one affected organs are liver and placenta (Schmidt et al.
highly-potent and selective inhibitor of Bcl-2 family, 1995). Liver of this mice shows defective structure
Navitoclax, caused durable single-agent response in with enlarged sinusoidal space and dissociated
heavily pretreated patients (Gandhi et al. 2011). parenchymal, frequently apoptotic cells. Other fun-
There are a lot of controversy reports according to damental embryogenetic process such as muscle
the prognostic role of Bcl-2 in lung carcinoma patients. development (Bladt et al. 1995), nervous system
One meta-analysis, including 25 trials with totaly 3370 formation, bone remodeling and angiogenesis are
participants, shows that patients with Bcl-2-positive highly affected by MET (Maina and Klein 1999).
tumors had significantly better survival than those with During the organogenesis c-MET transcripts are
Bcl-2-negative tumors (Martin et al. 2003). In the same detected in epithelia of lung, kidney and intestine while
meta-analysis, in case of patients with SCLC, data on HGF/SF1 is expressed in neighborhood mesenchyme
Bcl-2 expression were insufficient to give an definitive (Schmidt et al. 1995). In the adult tissue, the most
conclusion if Bcl-2 expression may be used as a important role of HGF/SF is regulation of response to
prognostic marker in this patients population. tissue damage. The rising serum level of circulating
Specific inhibition of Bcl-2 with Navitoclax has a HGF/SF1 and increased HGF/SF expression are
promising activity in pre-treated patients with SCLC observed after liver, kidney, heart or injury of other
and future clinical development should clarify if organs (Michalopoulos and DeFrances 1997; Matsum-
this inhibition may have a place in routine clinical oto and Nakamura 2001; Nakamura et al. 2000).
treatment of SCLC. In carcinogenesis, HGF/Met signaling plays
important role, especially in tumor invasiveness and
metastatic spread. Activation of MET in tumors can
9 MET occur most frequently through ligand-dependent
autocrine or paracrine mechanisms. Other oncogene,
c-MET is a proto-oncogene which encodes the tyrosine such as activated RAS can induce MET overexpres-
kinase receptor for HGF/SF1 (hepatocyte growth fac- sion (Mazzone and Comoglio 2006). Deregulation of
tor/scatter factor). Structurally, HGF/SF is a large, the HGF/MET signaling pathway may occur through
multi-domain protein showing structural similarity different mechanisms, including HGF and/or MET
with plasminogen, the proenzyme of plasmin, whose overexpression, MET gene amplification, mutations
primary role is the degradation of fibrin in the vascu- or rearrangements (Birchmeier et al. 2003).
lature. HGF/SF consists of six domains: an amino- Strongly MET expression was observed in 60%
terminal domain (N), four KRINGLE DOMAINS NSCLC and 25% SCLC (Ma et al. 2005, 2007).
(K1–K4) and a serine proteinase homology (SPH) Despite the rapid clinical response in NSCLC
domain, which lacks enzymatic activity as a result of patient with EGFR exon 19 mutant treated with
mutations in essential residues. MET kinase catalytic Gefitinib or Erlotinib, the vast majority of them will
activity is induced upon MET activation by HGF and it develop resistance to quinozoline-based drug treat-
is responsible for transphosphorylation of the tyrosines ment. By approximately 50% of patients reason to
Molecular Biology and Genetics of Lung Cancer 11

therapy failure is occurrence of a secondary mutation dual EGFR/MET inhibition was observed in MET
in EGFR (T790M) (Kosaka et al. 2006). FISH ? NSCLC as well as in FISH-/IHC+.
It has been reported that resistance to gefitinib in Targeting EGFR with gefitinib or erlotinib and
one gefitinib-sensitive EGFR exon 19 mutant NSCLC MET with specific inhibitors may represent a prom-
HCC827 lung cancer cell-line was caused by focal ising strategy to treat lung cancer. This new thera-
amplification of the MET proto-oncogene (Engelman peutic approach already reveals promising response in
et al. 2007). In this cell line, where ERBB3/PI3K/Akt patients with lung carcinoma.
signaling pathway is required for gefitinib induced
apoptosis, resistance was induced exposing these cells
to increasing concentrations of this drug for 6 months. 10 EGFR
This resistance cell-line was known as HCC827 GR
and in comparison to parental cell-line it showed The epidermal growth factor receptor (EGFR; ErbB-1)
marked focal amplification within chromosome is the cell-surface receptor which belongs together with
7q31.1–7q33.3, which contains the MET proto- other three closely related receptor tyrosine kinases—
oncogene. In this cell-line increased MET amplifica- HER2/c-neu (ErbB-2), Her 3 (ErbB-3) and Her 4
tion (5–10 times) was observed and sequencing ana- (ErbB-4) to family of ErbB family of receptors. EGFR
lysis provided no evidence of mutations in MET. The is a monomeric 170-kD transmembrane glycoprotein
treatment of resistant cells with specific MET tyrosine which consists of an extracellular ligand-binding
kinase inhibitor, PHA-665752 in the combination domain, a transmembrane lipophilic segment and an
with gefitinib-caused growth inhibition and induced intracellular protein kinase domain with the regulatory
apoptosis. In the same cells, gefitinib alone markedly COOH-terminal segment. The receptor exists as an
reduced phosphorylation of EGFR, but without sig- inactive monomer but after ligand-binding (epidermal
nificantly effects ERBB3 and Akt phosphorylation. In growth factor, transforming growth factor a(TGFa),
contrast, gefitinib in combination with PHA-665752 Heparin-binding EGF-like growth factor, betacellulin,
completely suppressed ERBB3 and Akt phosphor- amphiregulin), EGFR dimerization occurs. It results in
ylation in these cells, suggesting that observed resis- activation of the intracellular domain of receptor and
tance to gefitinib is a consequence of increased autophosphorylation of several tyrosine residues in the
MET signaling. In other experiments it has been C-terminal domain of EGFR. These events lead to the
found that MET amplification caused continuing initiation of a cascade of biochemical intracellular
activation of PI3K/Akt signaling in the presence of events that are involved in the mitogenic signal trans-
gefitinib by maintaining ERBB3 phosphorylation. duction of malignant cell. Phosphorylated tyrosine
To evaluate clinical relevance of these findings, 18 residues play important role as a docking site for pro-
tumors of patients with NSCLC, who initially teins SH2, Grb2 and Shc and enzymes such as phos-
responded to treatment with gefitinib or erlotinib, but pholipase C, phosphatidylinositol-3 kinase (PI-3K)
after some period showed tumor regrowth, were and the Src family kinases. These proteins are impor-
analyzed for MET copy status. MET amplification tant for initiation of multiple intracellular signal
was detected in 4 out of 18 (22%) gefitinib/erlotinib- transduction cascades that cause activation of many
resistant tumor specimens. Notably, dual inhibition of downstream pathways of EGFR such as ras/mitogen-
Met/EGFR was investigated in an globally random- activated protein kinase (MAPK), PI-3K/Akt and sig-
ized, double-blind phase II study (OAM4558g) nal transducer and activator of transcription (STAT)
(Spigel et al. 2011). One hundred and twenty-eight pathways (Hynes and Lane 2005).
NSCLC patients were equally randomized to receive In normal embryologyical development, EGFR
MetMAb, a monovalent monoclonal antibody that plays important role in development of lungs, eyes,
specifically binds the Met receptor, plus erlotinib or epidermis, hair and central nervous system (Liu and
placebo plus erlotinib in second/third line treatment. Neufeld 2007). The lungs from EGFR mutant mice
It has been found that addition of MetMab to erlotinib showed undifferentiated epithelium in the respira-
in these patients significantly improved PFS and OS tory bronchioles and alveoli and increased amounts
(12.6 vs. 3.8 months). The toxicity was similar in of cells in the alveolar septae (Sibilia and Wagner
both treatment arms. An overall survival benefit from 1995).
12 D. Milanovic

EGFR is overexpressed in 50% of NSCLC; the also be reason for gefitinib resistance (Linardou et al.
highest rate (80%) is detected in squamous cell car- 2008; Pratilas et al. 2008). As described previously,
cinoma (Meert et al. 2002). translocations in ALK, MET amplification and
EGFR amplification is detected in ca. 30% of increased expression of HGF may contribute to gefi-
patients with squamous cell and 15% with ADC of tinib resistance in NSCLC with wild-type EGFR.
lungs. EGFR kinase domain mutations may be found For some patients with EGFR mutations, treatment
in approximately 40% of Asian patients while in the with small tyrosin kinase inhibitors represents a valu-
USA, these mutations are detected in ca. 10% in able alternative for conventional chemotherapy or new
tumor specimens. In SCLC, amplification and kinase therapeutic option in the case of therapy failure.
domain mutations are very rare events. The most To expand therapeutical window of EGFR inhibition,
frequently occurring EGFR kinase domain mutations the complex molecular mechanism which caused
(*80%) are in-frame deletions within exon 19 resistance, should be better understood. Also the
(Godin-Heymann et al. 2007) or the L858R mutant combination with other targeted therapies may play
within exon 21 (point mutation) (Mulloy et al. 2007) important role in treatment of lung cancer in the future.
which occur most frequently in women, nonsmokers
and ADC histology. The most dramatic clinical
response to EGFR targeted therapies with small 11 p53
molecule tyrosine kinase inhibitors such as Gefitinib
or Erlotinib, occur in patients which tumors harboring p53 is a protein that in humans is encoded by the
these mutations (Pao et al. 2004). On the other hand, TP53 gene (Matlashewski et al. 1984). p53 plays
some of EGFR mutations, which occur in exons important role in apoptosis, genomic stability and
18–21 are associated with resistance to small mole- inhibition of angiogenesis. In multi-cellular organism
cule EGFR inhibitors (Wu et al. 2008). In *5% of it is acting as a tumor suppressor by activation of
patients with NSCLC small insertions or duplications DNA repair, induction of growth arrest by holding the
in exon 20 may be detected. Consisting with in vitro cell cycle at the G1/S regulation point on DNA
data, which showed that such mutations are less damage recognition and initiation of apoptosis, in
sensitive to EGFR TKIs, patients with tumors har- case of irreparable DNA damage. Because of its
boring exon 20 insertions show progressive disease suppressor role, p53 has been described as ‘‘the
during therapy with EGFR TKIs (Wu et al. 2008). guardian of the genome’’. In case of TP 53 mutation,
Despite the early response to EGFR TKIs, most of tumor suppression is severely impaired (Millau et al.
patients with exon 19 and exon 21 mutations will 2008). TP 53 mutations can be found in 60–75% of
progress. This acquired resistance may be caused by lung cancer including both NSCLC and SCLC (Oli-
development of T790M mutation, which is encoded vier et al. 2002). Majority of mutations are missense
by exon 20 (Inukai et al. 2006), L747 S (exon 19) mutations in exons 5–8 (Bodner et al. 1992).
(Costa et al. 2008), D761Y (exon 19) (Balak et al.
2006) and T854A (exon 21) (Bean et al. 2008). On the
other hand, different genetic changes, such as muta- 12 Other Factors
tions in PIK3CA (Kawano et al. 2006), crosstalk
between EGFR and insulin-like growth factor recep- Cyclooxygenase-2 (COX2) and COX1 are two COX
tor 1 (Sharma et al. 2010), loss-of PTEN expression enzymes which are responsible for the conversion of
(Sos et al. 2009) may cause resistance to EGFR TKIs arachidonic acid to prostaglandins (PGs) and other
in spite of present drug-sensitive EGFR mutations. bioactive lipids. COX2 plays key role in inflammatory
Tumors, with wild type EGFR, are generally resistant response and it has been found that inflammation may
to gefitinib treatment (Mok et al. 2009). Somatic have tumor promoting effects. COX2 may be upregu-
mutations in other genes may be reason for this drug lated with different stimuli such as interleukin (IL)-1,
insensitivity. For example, activating mutations of tumor necrosis factor a, platelet-derived growth factor
KRAS (codons 12 and 13 in GTPase domain) are and epidermal growth factor. In neoplastic disease,
detected in 15–25% of NSCLC or BRAF mutations, COX2 may play important role in angiogenesis,
which are detected in 2–3% NSCLC specimens, may apoptosis and tumor invasiveness. This enzyme is
Molecular Biology and Genetics of Lung Cancer 13

frequently overexpressed in NSCLC (*70%) while in obtained in last years, combined with synthetic
SCLC overexpression is not detected. Elevated COX2 lethality strategies may provide a new challenge to
protein levels and increased mRNA levels were asso- develop relevant in vitro and in vivo model for
ciated with poor prognosis of patients with NSCLC both of main histological entities of lung carcinoma.
(Wolff et al. 1998). Preclinical data showed that treat- Relevant model will open new therapeutic windows
ment of mice with selective COX2 inhibitor celecoxib, for testing new drug candidates which should be used
as a sole compound (Fulzele et al. 2006) or in the for treating these, until now, refractory disease.
combination with docetaxel may significantly inhibit
growth of A549 lung tumor xenografts (Shaik et al.
2006). References
In one double-blind randomized clinical phase III
trial (CYCLUS study) 319 patients were randomized Allen TD, Zhu CQ, Jones KD et al (2011) Interaction between
to receive celecoxib 400 mg or placebo in addition to MYC and MCL1 in the genesis and outcome of non-small-cell
palliative chemotherapy. This study failed to dem- lung cancer. Cancer Res 71:2212–2221
Amati B, Brooks MW, Levy N (1993) Oncogenic activity of
onstrate survival benefit in patients who were treated the c-Myc protein requires dimerization with Max. Cell
with celecoxib and chemotherapy (Koch et al. 2011). 72:233–245
PI3K/AKT/mTOR pathway is an intracellular Balak MN, Gong Y, Riely GJ et al (2006) Novel D761Y and
signaling pathway which is involved in the regulation common secondary T790M mutations in epidermal growth
factor receptor-mutant lung adenocarcinomas with acquired
of a number of cellular processes, such as transcrip- resistance to kinase inhibitors. Clin Cancer Res 12:6494–6501
tion, migration, angiogenesis, cell growth, prolifera- Barbie DA, Tamayo P, Boehm JS et al (2009) Systematic RNA
tion, apoptosis and glucose metabolism. PI3K is interference reveals that oncogenic KRAS-driven cancers
activated by several hormones including insulin, require TBK1. Nature 2009 462:108–112
Bean J, Riely GJ, Balak M et al (2008) Acquired resistance to
growth factors such as EGF, IGF, PDGF, NGF and epidermal growth factor receptor kinase inhibitors associ-
HGF by signals derived from receptors for extracel- ated with a novel T854A mutation in a patient with EGFR-
lular matrix molecules such as integrins, by several mutant lung adenocarcinoma. Clin Cancer Res 2008 14(22):
forms of cellular stress such as oxidative stress or cell 7519–7525
Beggs AD, Latchford AR, Vasen HF et al (2010) Peutz–Jeghers
swelling, and by activation of Ras. Akt is constitu- syndrome: a systematic review and recommendations for
tively activated at high rates in both NSCLC and management. Gut 59:975–986
SCLC (65%) (Brognard et al. 2001). Berghmans T, Paesmans M, Mascaux C et al (2006) Thyroid
The p16INK4a is a tumor suppressor protein acting transcription factor 1-a new prognostic factor in lung
cancer: a meta-analysis. Ann Oncol 17:1673–1676
as an inhibitor of CDK4 and CDK6, the D-type cyclin- Birchmeier C, Birchmeier W, Gherardi E et al (2003) Met,
dependent kinases which are responsible for initiation metastasis, motility and more. Nat Rev Mol Cell Biol 4:
of phosphorylation of the retinoblastoma tumor sup- 915–925
pressor protein, RB, which is encoded by the RB1 gene Bladt F, Riethmacher D, Isenmann S et al (1995) Essential role
for the c-met receptor in the migration of myogenic
located on chromosome 13q14.1–q14.2. RB1 expres- precursor cells into the limb bud. Nature 376:768–771
sion is deregulated in 15–30% of NSCLC and[90% of Bodner SM, Minna J, Jensen SM et al (1992) Expression of
SCLC, but without prognostic significance (Shimizu mutant p53 proteins in lung cancer correlates with the class
et al. 1994). of p53 gene mutation. Oncogene 7:743–749
Brognard J, Clark AS, Ni Y et al (2001) Akt/protein kinase B is
Loss-of heterozygosity (LOH) in 3p is also com- constitutively active in non-small cell lung cancer cells and
mon genetic alteration detected in almost all types of promotes cellular survival and resistance to chemotherapy
lung cancer (Wistuba et al. 2000), but its contribution and radiation. Cancer Res 61:3986–3997
to lung cancer development is yet to be clarified. Chao DT, Korsmeyer SJ (1998) BCL-2 family: regulators of
cell death. Annu Rev Immunol 16:395–419
Choi YL, Soda M, Yamashita Y, Ueno T et al (2010) EML4-
ALK mutations in lung cancer that confer resistance to ALK
13 Conclusion inhibitors. N Engl J Med 363:1734–1739
Costa DB, Schumer ST, Tenen DG et al (2008) Differential
responses to erlotinib in epidermal growth factor receptor
Despite the intensive research and development, (EGFR)-mutated lung cancers with acquired resistance to
prognosis of patients with inoperable NSCLC and gefitinib carrying the L747S or T790M secondary mutations.
SCLC remain poor. Microarray data, which are J Clin Oncol 26:1182–1184
14 D. Milanovic

Davis AC, Wims M, Spotts GD et al (1993) A null c-myc III trial (CYCLUS study) by the Swedish Lung Cancer
mutation causes lethality before 10.5 days of gestation in Study Group. Eur J Cancer 47:1546–1555
homozygotes and reduced fertility in heterozygous female Koivunen JP, Mermel C, Zejnullahu K et al (2008) EML4-ALK
mice. Genes Dev 7:671–682 fusion gene and efficacy of an ALK kinase inhibitor in lung
Downward J (2003) Targeting RAS signalling pathways in cancer. Clin Cancer Res 14:4275–4283
cancer therapy. Nat Rev Cancer 3:11–22 Kosaka T, Yatabe Y, Endoh H et al (2006) Analysis of
Engelman JA, Zejnullahu K, Mitsudomi T et al (2007) MET epidermal growth factor receptor gene mutation in patients
amplification leads to gefitinib resistance in lung cancer by with non-small cell lung cancer and acquired resistance to
activating ERBB3 signaling. Science 316:1039–1043 gefitinib. Clin Cancer Res 12(19):5764–5769
Fulzele SV, Chatterjee A, Shaik MS et al (2006) Inhalation Kwak EL, Bang YJ, Camidge DR et al (2010) Anaplastic
delivery and anti-tumor activity of celecoxib in human lymphoma kinase inhibition in non-small-cell lung cancer.
orthotopic non-small cell lung cancer xenograft model. N Engl J Med 363:1693–1703
Pharm Res: 2094–2106 Kwei KA, Kim YH, Girard L et al (2008) Genomic profiling
Gandhi L, Camidge DR, Ribeiro de Oliveira M et al (2011) identifies TITF1 as a lineage-specific oncogene amplified in
Phase I study of Navitoclax (ABT-263), a novel Bcl-2 lung cancer. Oncogene 27:3635–3640
family inhibitor, in patients with small-cell lung cancer and Linardou H, Dahabreh IJ, Kanaloupiti D et al (2008) Assessment
other solid tumors. J Clin Oncol 29:909–916 of somatic k-RAS mutations as a mechanism associated with
Garraway LA, Sellers WR (2006) Lineage dependency and resistance to EGFR-targeted agents: a systematic review and
lineage-survival oncogenes in human cancer. Nat Rev meta-analysis of studies in advanced non-small-cell lung
Cancer 6:593–602 cancer and metastatic colorectal cancer. Lancet Oncol 9:
Godin-Heymann N, Bryant I, Rivera MN et al (2007) 962–972
Oncogenic activity of epidermal growth factor receptor Liu B, Neufeld AH (2007) Activation of epidermal growth
kinase mutant alleles is enhanced by the T790M drug factor receptors in astrocytes: from development to neural
resistance mutation. Cancer Res 67:7319–7326 injury. J Neurosci Res 85:3523–3529
Guazzi S, Price M, De Felice M et al (1990) Thyroid nuclear Loriot Y, Mordant P, Deutsch E (2009) Are RAS mutations
factor 1 (TTF-1) contains a homeodomain and displays a predictive markers of resistance to standard chemotherapy?
novel DNA binding specificity. EMBO J 9:3631–3639 Nat Rev Clin Oncol 6:528–534
Hynes NE, Lane HA (2005) ERBB receptors and cancer: the Ma PC, Jagadeeswaran R, Jagadeesh S et al (2005) Functional
complexity of targeted inhibitors. Nat Rev Cancer 5: expression and mutations of c-Met and its therapeutic
341–354 inhibition with SU11274 and small interfering RNA in
Inukai M, Toyooka S, Ito S et al (2006) Presence of epidermal non-small cell lung cancer. Cancer Res 65:1479–1488
growth factor receptor gene T790M mutation as a minor Ma PC, Tretiakova MS, Nallasura V et al (2007) Downstream
clone in non-small cell lung cancer. Cancer Res 66: signalling and specific inhibition of c-MET/HGF pathway in
7854–7858 small cell lung cancer: implications for tumour invasion.
Iwakawa R, Kohno T, Kato M et al (2011) MYC amplification Br J Cancer 97:368–377
as a prognostic marker of early-stage lung adenocarcinoma Maina F, Klein R (1999) Hepatocyte growth factor, a versatile
identified by whole genome copy number analysis. Clin signal for developing neurons. Nat Neurosci 2:213–217
Cancer Res 17:1481–1489 Martin B, Paesmans M, Berghmans T et al (2003) Role of
Jansen M, Ten Klooster JP, Offerhaus GJ et al (2009) LKB1 Bcl-2 as a prognostic factor for survival in lung cancer: a
and AMPK family signaling: the intimate link between cell systematic review of the literature with meta-analysis.
polarity and energy metabolism. Physiol Rev 89:777–798 Br J Cancer 89:55–64
Ji H, Ramsey MR, Hayes DN, Fan C et al (2007) LKB1 Mascaux C, Iannino N, Martin B et al (2005) The role of RAS
modulates lung cancer differentiation and metastasis. Nature oncogene in survival of patients with lung cancer: a system-
448:807–810 atic review of the literature with meta-analysis. Br J Cancer
Jiang SX, Sato Y, Kuwao S et al (1995) Expression of bcl-2 92:131–139
oncogene protein is prevalent in small cell lung carcinomas. Matlashewski G, Lamb P, Pim D et al (1984) Isolation and
J Pathol 177:135–138 characterization of a human p53 cDNA clone: expression of
Katayama R, Khan TM, Benes C et al (2011) Therapeutic the human p53 gene. EMBO J 3:3257–3262
strategies to overcome crizotinib resistance in non-small Matsumoto K, Nakamura T (2001) Hepatocyte growth factor:
cell lung cancers harboring the fusion oncogene EML4- renotropic role and potential therapeutics for renal diseases.
ALK. Proc Natl Acad Sci U S A 108:7535–7540 Kidney Int 59:2023–2038
Kawano O, Sasaki H, Endo K et al (2006) PIK3CA mutation status Matsumoto S, Iwakawa R, Takahashi K et al (2007) Prevalence
in Japanese lung cancer patients. Lung Cancer 54:209–215 and specificity of LKB1 genetic alterations in lung cancers.
Kimura S, Hara Y, Pineau T et al (1996) The T/ebp null mouse: Oncogene 26(40):5911–5918
thyroid-specific enhancer-binding protein is essential for the Mazzone M, Comoglio PM (2006) The Met pathway: master
organogenesis of the thyroid, lung, ventral forebrain, and switch and drug target in cancer progression. FASEB J
pituitary. Genes Dev 10: 60–69 20(10):1611–1621
Koch A, Bergman B, Holmberg E et al (2011) Effect of Meert AP, Martin B, Delmotte P et al (2002) The role of EGF-R
celecoxib on survival in patients with advanced non-small expression on patient survival in lung cancer: a systematic
cell lung cancer: a double blind randomised clinical phase review with meta-analysis. Eur Respir J 20:975–981
Molecular Biology and Genetics of Lung Cancer 15

Michalopoulos GK, DeFrances MC (1997) Liver regeneration. Soucek L, Whitfield J, Martins CP et al (2008) Modelling Myc
Science 276:60–66 inhibition as a cancer therapy. Nature 455:679–683
Millau JF, Bastien N, Drouin R (2008) P53 transcriptional Spigel DR, Ervin TJ, Ramlau R et al (2011) Final efficacy results
activities: a general overview and some thoughts. Mutat Res from OAM4558g, a randomized phase II study evaluating
681:118–133 MetMAb or placebo in combination with erlotinib in
Mok TS, Wu YL, Thongprasert S et al (2009) Gefitinib or advanced NSCLC. J Clin Oncol 29:(suppl; abstr 7505)
carboplatin–paclitaxel in pulmonary adenocarcinoma. Sunaga N, Imai H, Shimizu K et al (2011) Oncogenic KRAS-
N Engl J Med 361(10):947–957 induced interleukin-8 overexpression promotes cell growth
Mulloy R, Ferrand A, Kim Y et al (2007) Epidermal growth and migration and contributes to aggressive phenotypes of
factor receptor mutants from human lung cancers exhibit non-small cell lung cancer. Int J Cancer (Epub ahead of print:
enhanced catalytic activity and increased sensitivity to 4 May, 2011)
gefitinib. Cancer Res 67(5):2325–2330 Talbot SG, Estilo C, Maghami E et al (2005) Gene expression
Nakamura T, Mizuno S, Matsumoto K et al (2000) Myocardial profiling allows distinction between primary and metastatic
protection from ischemia/reperfusion injury by endogenous squamous cell carcinomas in the lung. Cancer Res 65:
and exogenous HGF. J Clin Invest 106:1511–1519 3063–3071
Olivier M, Eeles R, Hollstein M et al (2002) The IARC TP53 Tanaka H, Yanagisawa K, Shinjo K et al (2007) Lineage-
database: new online mutation analysis and recommenda- specific dependency of lung adenocarcinomas on the lung
tions to users. Hum Mutat 19:607–614 development regulator TTF-1. Cancer Res 67:6007–6011
Pao W, Miller V, Zakowski M, Doherty J et al (2004) EGF Vakiani E, Solit DB (2011) KRAS and BRAF: drug targets and
receptor gene mutations are common in lung cancers from predictive biomarkers. J Pathol 223:219–229
‘‘never smokers’’ and are associated with sensitivity of Vennstrom B, Sheiness D, Zabielski J et al (1982) Isolation and
tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A characterization of c-myc, a cellular homolog of the
101(36):13306–13311 oncogene (v-myc) of avian myelocytomatosis virus strain
Pelengaris S, Khan M, Evan G (2002) c-MYC: more than just a 29. J Virol 42:773–779
matter of life and death. Nat Rev Cancer 2:764–776 Voortman J, Lee JH, Killian JK et al (2010) Array comparative
Pezzella F, Turley H, Kuzu I et al (1993) bcl-2 protein in non- genomic hybridization-based characterization of genetic
small-cell lung carcinoma. N Engl J Med 329:690–694 alterations in pulmonary neuroendocrine tumors. Proc Natl
Pratilas CA, Hanrahan AJ, Halilovic E et al (2008) Genetic Acad Sci U S A 107(29):13040–13045
predictors of MEK dependence in non-small cell lung Wagner PL, Stiedl AC, Wilbertz T et al (2011) Frequency and
cancer. Cancer Res 68:9375–9383 clinicopathologic correlates of KRAS amplification in non-
Salgia R, Skarin AT (1998) Molecular abnormalities in lung small cell lung carcinoma. Lung Cancer (Epub ahead of print:
cancer. J Clin Oncol 16:1207–1217 7 Apr, 2011)
Sanchez-Cespedes M, Parrella P, Esteller M et al (2002) Weir BA, Woo MS, Getz G et al (2007) Characterizing the cancer
Inactivation of LKB1/STK11 is a common event in genome in lung adenocarcinoma. Nature 450: 893–398
adenocarcinomas of the lung. Cancer Res 62:3659–3662 Winslow MM, Dayton TL, Verhaak RG et al (2011) Suppres-
Schmidt C, Bladt F, Goedecke S et al (1995) Scatter factor/ sion of lung adenocarcinoma progression by Nkx2-1. Nature
hepatocyte growth factor is essential for liver development. 473:101–104
Nature 373:699–702 Wistuba I, Behrens C, Virmani AK et al (2000) High resolution
Shaik MS, Chatterjee A, Jackson T et al (2006) Enhancement chromosome 3p allelotyping of human lung cancer and
of antitumor activity of docetaxel by celecoxib in lung preneoplastic/preinvasive bronchial epithelium reveals mul-
tumors. Int J Cancer 118:396–404 tiple, discontinuous sites of 3p allele loss and three regions
Sharma SV, Lee DY, Li B et al (2010) A chromatin-mediated of frequent breakpoints. Cancer Res 60:1949–1960
reversible drug-tolerant state in cancer cell subpopulations. Wolff H, Saukkonen K, Anttila S et al (1998) Expression of
Cell 141:69–80 cyclooxygenase-2 in human lung carcinoma. Cancer Res
Shimizu E, Coxon A, Otterson GA et al (1994) RB protein 58:4997–5001
status and clinical correlation from 171 cell lines repre- Wong DW, Leung EL, So KK et al (2009) The EML4-ALK
senting lung cancer, extrapulmonary small cell carcinoma, fusion gene is involved in various histologic types of lung
and mesothelioma. Oncogene 9:2441–2448 cancers from nonsmokers with wild-type EGFR and KRAS.
Sibilia M, Wagner EF (1995) Strain-dependent epithelial defects Cancer 115(8):1723–1733
in mice lacking the EGF receptor. Science 269:234–238 Wu JY, Wu SG, Yang CH et al (2008) Lung cancer with
Soda M, Choi YL, Enomoto M, Takada S et al (2007) epidermal growth factor receptor exon 20 mutations is
Identification of the transforming EML4-ALK fusion gene associated with poor gefitinib treatment response. Clin
in non-small-cell lung cancer. Nature 448:561–566 Cancer Res 14:4877–4882
Soda M, Takada S, Takeuchi K et al (2008) A mouse model Yatabe Y, Mitsudomi T, Takahashi T (2002) TTF-1 expression in
for EML4-ALK-positive lung cancer. Proc Natl Acad Sci pulmonary adenocarcinomas. Am J Surg Pathol 26:767–773
U S A 105:19893–19897 Zhao X, Weir BA, LaFramboise T et al (2005) Homozygous
Sos ML, Koker M, Weir BA et al (2009) PTEN loss contributes deletions and chromosome amplifications in human lung
to erlotinib resistance in EGFR-mutant lung cancer by carcinomas revealed by single nucleotide polymorphism
activation of Akt and EGFR. Cancer Res 69:3256–3261 array analysis. Cancer Res 65:5561–5570
Angiogenesis and Lung Cancer
Wenyin Shi and Dietmar W. Siemann

Contents Abstract
Lung cancer is one of the most frequent causes of
1 Introduction.............................................................. 17 cancer deaths worldwide. Current treatment regi-
2 Angiogenesis ............................................................. 18 mens with conventional anticancer therapies offer
only a limited survival benefit. There clearly exists
3 Angiogenesis in Lung Cancer ................................ 19
a need for the development of new therapeutic
4 Anti-Angiogenic Therapy........................................ 23 strategies. Recent evidence suggests that angio-
4.1 Drugs that Block Angiogenic Factors ...................... 24
genesis is critical to lung cancer progression and
4.2 Drugs that Inhibit Endothelial Cell Function ........... 30
4.3 Drugs that Block Breakdown related to poor prognosis. Consequently tumor
of Extracellular Matrix.............................................. 31 angiogenesis, a process that features an imbalance
4.4 Drugs that Target Survival Factors of Neovessels... 31 between pro and antiangiogenic mediators, is
5 Vascular Disrupting Therapies .............................. 32 being targeted by novel therapies in the treatment
6 Conclusions ............................................................... 33
of lung cancer. A variety of therapeutic approaches
and agents have been developed to compromise the
References.......................................................................... 33
growth and/or function of tumor vasculature. In
October 2006, bevacizumab was the first antian-
giogenic agent approved by the US Food and Drug
Administration for the treatment of advanced,
nonsquamous, nonsmall cell lung cancer in com-
bination with platinum-based chemotherapy. Other
antiangiogenic agents are actively being evaluated
in pre-clinical and clinical settings.

1 Introduction

Lung cancer is a significant public health problem in


W. Shi (&)
Department of Radiation Oncology, the United States and the world. In the United States,
Thomas Jefferson University, lung cancer ranks as the second most common cancer
111 S 11 ST Suite G301, among both men and women. In 2010, an estimated
Philadelphia, PA 19107, USA
222,520 new cases of lung cancer were diagnosed in
e-mail: wenyin.shi@jefferson.edu
the United States; a number representing approxi-
D. W. Siemann
mately 14.6% of all new cancers diagnosed. Lung
Department of Radiation Oncology,
Shands Cancer Center, University of Florida, cancer is the most common cause of cancer-related
Gainesville, FL 32610, USA deaths in both female and male, accounting for

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 17
DOI: 10.1007/174_2011_260, Ó Springer-Verlag Berlin Heidelberg 2011
18 W. Shi and D. W. Siemann

157,300 deaths in 2010 (Jemal et al. 2010). Although


the incidence of lung cancer is now declining in men, 2 Angiogenesis
the incidence in women continues to increase (Weir
et al. 2003), probably due to changing smoking habits. Angiogenesis is a process that allows the develop-
According to World Health Organization histo- ment and formation of new blood vessels from a pre-
logic classification schemes (WHO 1979), there are existing vascular network. This process is complex
four primary pathological types of lung cancer: small- and involves multiple sequential, interactive steps as
cell carcinoma, squamous-cell carcinoma, adenocar- well as a variety of cells, soluble factors, and the
cinoma and large-cell carcinoma. However, for ther- extracellular matrix. The sequential steps include:
apeutic purposes, lung cancer is generally divided into degradation of basement membranes, migration and
small-cell lung cancer (SCLC) or non-small-cell lung proliferation of endothelial cells, lumen formation,
cancer (NSCLC) with the latter representing approx- and stabilization of neovessels. Under physiological
imately 80% of all lung cancer patients (Edwards circumstances, angiogenesis is a rare event in adults,
et al. 2005). occurring almost exclusively in the female repro-
Although prevention and early detection are critical duction system (Folkman 1995; Risau 1997). It is
to improve treatment outcomes, these have proven normally suppressed and observed only transiently.
difficult in lung cancer. A major reason is that only However, angiogenesis can be activated in response
approximately 15% of lung cancers are discovered to tissue damage, and it is associated with a variety of
while still localized. Local treatment for early-stage pathological conditions including cancer (Folkman
disease, particularly surgical interventions, can 2002). While angiogenesis in itself is not sufficient for
improve patient survival, yet less than 50% of patients continued tumor growth, its absence severely com-
are cured, principally due to the presence of undetected promises or halts the expansion of a tumor cell pop-
occult local or metastatic disease (Mountain and Her- ulation. Indeed, it is believed that tumors can not
mes 2003; Posther and Harpole 2006). Radiotherapy grow to a size larger than a few cubic mm without
and chemotherapy typically are applied in more initiating the angiogenic process (Folkman 1971,
advanced disease. Still, survival in patients with lung 1975, 2002). Furthermore, there is evidence that
cancer remains poor. The 5 year survival rate for all angiogenesis may be present in pre-malignant lesions
stages combined is only 5–15% (Comis 2003; Kepka such as epithelial dysplasia even prior to development
et al. 2009). The majority of patients die from disease of invasive cancer (Fontanini et al. 1995; Keith et al.
progression locally, at distant sites, or both. 2000).
Pathologic staging, which incorporates factors A balance of pro- and anti-angiogenic factors
such as tumor size and grade, nodal status and pres- carefully regulates the angiogenic potential of endo-
ence or absence of distant metastases, provides the thelial cells. While tightly controlled under normal
best prediction of treatment outcome (Mountain 2000; physiological conditions, this rigid control is absent in
Beadsmoore and Screaton 2003). However, because angiogenesis associated with tumors. Indeed altera-
the growth of primary tumors and metastases is tions of the expression and/or function of pro-angio-
angiogenesis dependent (Folkman 1971, 2002), a genic and anti-angiogenic molecules that disrupt the
great deal of attention has recently been paid to the normal balance appear to be responsible for tumor
role of this process not only in lung cancer formation, angiogenesis. The regulatory factors involved may
progression, and prognosis, but also in the develop- mediate any one of a cascade of steps in the process
ment of novel therapeutic strategies for this disease. of angiogenesis. As a consequence the characteristics
In October 2006, the anti-angiogenic agent bev- of endothelial cells and associated perivascular
acizumab was granted a labeling extension by the US structures (pericytes, vascular smooth muscle cells)
Food and Drug Administration for the first-line can be dramatically altered.
treatment of advanced, nonsquamous, NSCLC in Vascular endothelial growth factor (VEGF) is the
combination with platinum-based chemotherapy most potent and specific growth factor for endothelial
(Sandler et al. 2006). Many other agents that target cells. VEGF can increase vascular permeability,
tumor-associated vasculature are under development induce endothelial cell proliferation and migration,
and/or in various phases of clinical trial assessment. activate proteases for extra-cellular matrix
Angiogenesis and Lung Cancer 19

degradation, and inhibit apoptosis of endothelial cells and/or suppressing anti-angiogenic factors. The point
(Senger et al. 1986; Connolly et al. 1989; Watanabe at which angiogenesis is triggered by tumor cells is
and Dvorak 1997; Ferrara 2002). VEGF is comprised known as the angiogenic switch (Bergers and Benja-
of a family of five isoforms which bind with high min 2003). Tumor angiogenesis is a complex, well-
affinity to tyrosine kinase associated receptors that are orchestrated, cascade of steps, which initiated by
present on endothelial cells (Ferrara et al. 2003). VEGF induced vasodilation and increased vascular
Another class of endothelial cell specific molecules is permeability, extravasation of plasma proteins such as
the angiopoietin family. It includes at least four fibrinogen (Roodink and Leenders 2010), and the
members (angiopoietins 1–4) of which Ang-1 and Ang-2 mediated dissociation of pericytes from endo-
Ang-2 appear to be most relevant. Ang-1 and Ang-2 are thelial cells (Holash et al. 1999). The vascular base-
secreted proteins that interact bind the Tie-2 receptor ment membrane and extracellular matrix surround
with similar affinities but with opposing functional existing capillaries are then degraded by proteases
effects; Ang-1/Tie-2 signaling controls vessel quies- produced by tumor cells (Werb et al. 1999). Subse-
cence and stability while Ang-2/Tie-2 association quently, endothelial cells proliferate, invade the sur-
allows for vessel plasticity and destabilization (Papa- round extracellular matrix and form hollow tubes.
petropoulos et al. 1999). The Ang-2/Tie-2 interaction This process involves Delta-like ligand 4/Notch 1
sensitizes the vasculature to growth factors, such as induced differentiation (Roca and Adams 2007).
VEGF, to stimulate endothelial cells, angiogenesis and Finally, blood vessel sprouts fuse with other sprouts
vascular remodeling (Holash et al. 1999). to form vascular loops (Roodink and Leenders 2010;
In addition, there are numerous non-specific Ucuzian et al. 2010). Angiogenesis not only permits
angiogenic growth factors that can also affect endo- further growth of the tumor, but also provides a
thelial cells. These include platelet-derived growth pathway for migrating tumor cells to gain access to
factor (PDGF), basic fibroblast growth factor (bFGF/ the systemic circulation and eventually establish dis-
FGF-2), acidic fibroblast growth factor (aFGF/FGF- tant metastases (Folkman 1971, 1990b). Unlike blood
1), fibroblast growth factor-3 (FGF-3/int-2), fibroblast vessels in normal tissue, tumor-associated vasculature
growth factor-4 (FGF-4/hst/K-FGF), hepatocyte is irregular and unstable, likely due to the over-pro-
growth factor/scatter factor (HGF/SF), transforming duction of pro-angiogenic proteins such as VEGF
growth factor-a (TGF-a), transforming growth factor- (Bergers and Benjamin 2003). Tumor-associated
b (TGF-b), tumor necrosis factor-a (TNF-a), granu- blood vessels are excessively branched and chaotic.
locyte colony stimulating factor, interleukin-8, Targeting tumor vascular supply is widely recognized
pleiotropin, and angiogenin, to name just a few as an attractive anti-cancer strategy.
(Moore et al. 1998).
A growing number of endogenous anti-angiogenic
factors also been discovered. To date, these include 3 Angiogenesis in Lung Cancer
endostatin, angiostatin, vasostatin, interferon-a, b, c,
METH-1 and METH-2, antithrombin III, and VEGF The lungs are highly vascularized and highly depen-
inhibitor (Kerbel 2000, 2008; Gordon et al. 2010). dent on intact vasculature for efficient function.
These factors have great structural diversity and Endothelial cells lining the lumen surfaces of blood
activity. Some of the most notable, endostatin and vessels are not only a mechanical barrier but also play
angiostatin, are cleavage fragments of proteins that an essential role in the regulation of blood flow,
normally lack anti-angiogenic activity (O’Reilly et al. vascular permeability, angiogenesis, and metastasis
1994; O’Reilly 1997; Takahashi et al. 2010). Table 1 (Paku 1998; Tuder et al. 2001; Chouaib et al. 2010).
lists endogenous factors that stimulate and inhibit Endothelial cells from normal and tumor tissues not
angiogenesis. only differ phenotypically but also in their gene
Generally, solid tumors cannot grow beyond *1– expression profiles (St Croix et al. 2000). Moreover,
2 mm in diameter without developing their own blood significantly different expression profiles of angio-
vessel networks to supply nutrients and dispose of genic proteins have been observed between different
waste products (Folkman 1990a). Tumors trigger lung cancer types (Yamashita et al. 1999; Takase
angiogenesis by producing pro-angiogenic factors et al. 2010; Lopez-Campos et al. 2011).
20 W. Shi and D. W. Siemann

Table 1 Endogenous regulatory factors involved in angiogenesis


Pro-angiogenic Factors Anti-angiogenic Factors
Vascular endothelial growth factor (VEGF-A, B, C, D, E) Angiostatin
Placental growth factor Endostatin
Platelet-derived growth factor (PDGF) Vasostatin
Basic fibroblast growth factor (bFGF/FGF-2) Thrombospondin-1 and internal fragment
Acidic fibroblast growth factor (aFGF/FGF-1) Vascular endothelial growth factor inhibitor
Fibroblast growth factor-3 (FGF-3/int-2) Fragment of platelet factor-4
Fibroblast growth factor-4 (FGF-4/hst/K-FGF) Derivative of prolactin
Hepatocyte growth factor/Scatter factor (HGF/SF) Restin
Transforming growth factor-a (TGF-a) Proliferin-related protein
Transforming growth factor-b (TGF-b) SPARC cleavage product
Tumor necrosis factor-a (TNF-a) Osteopontin cleavage product
Granulocyte colony stimulating factor Interferon-a, Interferon-b
Interleukin-8 METH-1, METH-2
Pleiotropin Angiopoietin-2
Angiogenin Antithrombin III fragment
Proliferin Interferon-inducible protein-10
Matrix metalloproteinases (MMPs) Tissue inhibitors of metalloproteinases (TIMPs)
Angiopoietin-2 Prolactin
Prostaglandin E1 and E2 Interleukin 1, 6, 12
Thymidine phosphorylase (TP) VEGF soluble receptor
Platelet-derived endothelial cell growth factor (PD-ECGF) Dll4
Intergrin
Ephrin

Typically, angiogenesis in tumors has been asses- McCulloch et al. 1995; Mattern et al. 1996; Takahashi
sed indirectly by determining intratumoral microves- et al. 2010), there are important aspects of the process
sel density (MVD). Blood vessels are usually of angiogenesis that MVD does not reflect. For
immunostained with a pan-endothelial marker, such example, it does not measure the degree of vascular
as factor VIII-related antigen, and counted (Guidi heterogeneity across the tumor, or the functions of the
et al. 1994). More recently, markers with an increased microvasculature such as blood flow or extent of
ability to highlight the entire tumor vasculature tumor hypoxia.
(CD31, CD34) have replaced factor VIII-related Results from a number of clinical investigations
antigen as the most commonly used pan-endothelial now have indicated that increased MVD is associated
markers (Miettinen 1993; Hasan et al. 2002). An with a poor prognosis. Indeed MVD has been shown
international consensus on the methodology and cri- to be an independent prognostic factor in a variety of
teria for evaluation of MVD has been put forth tumor types, including breast, bladder, ovarian,
(Vermeulen et al. 1996). MVD is a measure of one prostatic, pancreatic, melanoma, colorectal, and gas-
feature of the tumor vasculature, the density of blood tric carcinoma (Toi et al. 1993; Dickinson et al. 1994;
vessels in the regions of tumor with the highest con- Tanigawa et al. 1996; Papamichael 2001; Bono et al.
centration of blood vessels, referred to as ‘hot spots’. 2002; Khan et al. 2002; Lee et al. 2002; Massi et al.
While there is evidence, accumulated over the past 2002; Gadducci et al. 2003; Herrmann et al. 2007;
10 years, that correlates this parameter with angio- Minardi et al. 2008; Mahzouni et al. 2010). Many
genic growth factor expression, tumor growth and the studies also have associated the peak vessel density as
occurrence of distant metastases (Weidner et al. 1993; measured by MVD with a poor prognosis in NSCLC
Angiogenesis and Lung Cancer 21

(Macchiarini et al. 1992; Yamazaki et al. 1994; tumor repeatedly. Secondly, while MVD reflects
Trivella et al. 2007; Guo et al. 2009; Medetoglu et al. some aspects of the angiogenic process, it may not be
2010; Yang et al. 2010; Anagnostou et al. 2011; Wu a measure of the relative dependence of a particular
et al. 2011). In addition, the incidence of node tumor on angiogenesis, and changes in MVD do not
involvement increased with MVD and MVD was an necessarily correlate with changes in tumor growth
independent variable associated with lymph node rate.
metastasis (Fontanini et al. 1995). The role of MVD The expression level of angiogenic factors, either
as a prognostic factor in locally advanced completely quantified within tumor tissue or after secretion into
resected NSCLC treated with postoperative radio- body fluids, provides another indirect measure of
therapy and chemotherapy also has been reported tumor angiogenesis. The latter approach is particu-
(Angeletti et al. 1996). Since, SCLC is rarely treated larly appealing as it provides a noninvasive means of
by surgery, this disease has not been as well studied. investigating tumor angiogenic activity with potential
Still, despite the paucity of information in this class of diagnostic and prognostic implications. A number of
tumors, it should be noted that data available suggest such studies have been reported for lung cancer
a similar correlation between MVD and prognosis for patients.
SCLC as has been reported in NSCLC (Fontanini In NSCLC a significant role of increased VEGF
et al. 2002; Lucchi et al. 2002). and a correlation of VEGF expression with poor
However, not all lung cancer investigations have prognosis have been found (Bonnesen et al. 2009;
demonstrated relationships between vessel density Carrillo de Santa Pau et al. 2009; Chen et al. 2010;
and outcome. For example, in several recent studies Feng et al. 2010; Lin et al. 2010; Rades et al. 2010a).
MVD failed to be a predictor for survival in NSCLC VEGF-receptor (KDR) expression by endothelial
(Apolinario et al. 1997; Chandrachud et al. 1997; cells has also been associated with poor prognosis in
Pastorino et al. 1997; Decaussin et al. 1999; Ma- NSCLC (Koukourakis et al. 2000; Seto et al. 2006). A
cluskey et al. 2000). A recent meta-analysis also similar association between VEGF expression and
concluded MVD does not seem to be a prognostic poor prognosis also was reported in SCLC (Ohta et al.
factor in patients with non-metastatic surgically 1996; Salven et al. 1998). In addition to determining
treated non-small-cell lung carcinoma (Trivella et al. tissue and tumor VEGF protein and mRNA expres-
2007). These apparently contradictory results may sion, it is also possible to measure VEGF concentra-
arise from differences in staining methods, tumor tions in body fluids. When this was done in lung
heterogeneity, and inter-observer variability. Inter- cancer patients, serum or plasma VEGF levels were
estingly, in tumors with an ‘‘alveolar pattern’’, where observed to increase with tumor stage progression
there is little parenchymal destruction and alveolar (Takigawa et al. 1998; Matsuyama et al. 2000;
septae are present, prognosis is worse than in tumors Tamura et al. 2002). Also, patients with elevated
showing an ‘‘angiogenic pattern’’ (Pezzella et al. serum or plasma VEGF levels at diagnosis had a
1997). This suggests that some lung cancers may be poorer response to therapy and worse survival (Salven
capable of co-opting the existing vascular bed thereby et al. 1998; Tamura et al. 2001). When measured in
relying less on new vessel formation. In this cir- bronchoalveolar lavage fluid, raised VEGF levels
cumstance MVD is unlikely to be of prognostic util- were noted in patients with advanced NSCLC before
ity. Also, while in general, MVD is an important and during treatment (Ohta et al. 2002). However,
prognostic indicator, it has not yet been shown to be a other studies failed to find a relationship between
useful measure for assessment of anti-angiogenic NSCLC prognosis and serum VEGF level (Brattstrom
treatments (Hlatky et al. 2002). There are a number of et al. 1998). This is perhaps not surprising, since there
potential reasons for this. Firstly, determination of are pitfalls in the measurement of circulating VEGF
treatment effect, rather than prediction of prognosis, levels. For example, platelets contain a large amount
requires serial measurement. Generally, only small of VEGF, and depending on how samples are han-
samples of tumor can be obtained in a serial manner. dled, varying amounts of platelet associated VEGF
Since MVD by definition measures the peak vessel may be released. Consequently the use of plasma
density, use of small samples may affect accuracy and rather than serum samples for measurement of VEGF
it is technically difficult to sample similar areas of has been recommended (Webb et al. 1998). Since,
22 W. Shi and D. W. Siemann

VEGF is one of the most potent and specific factors of reflects tumor vascular perfusion and angiogenesis
tumor angiogenesis, the clinical possibilities of uti- (Purdie et al. 2001; Tateishi et al. 2002; Yi et al. 2004;
lizing VEGF associated measurements as markers of Ng et al. 2009). It has been used to differentiate
tumor growth and/or response to therapy remains an between benign and malignant lung nodules (Swensen
area of intense interest, particularly for those thera- et al. 2000), prediction of clinical outcome (Goh et al.
pies that target the VEGF pathway (Drevs 2003; 2009; Park et al. 2009), and monitor antiangiogenic
Mihaylova et al. 2007; Dalaveris et al. 2009; Carrillo- therapy in NSCLC (Ng et al. 2007a, 2007b). Although
de Santa Pau et al. 2010). perfusion CT has a clear role in the assessment of
Basic FGF is another potent stimulator of angio- vascular response in early phase clinical trials, further
genesis that is often over-expressed in lung cancer validation studies are required before these measure-
patients (Guddo et al. 1999). Indeed high serum bFGF ments will be accepted as surrogate markers in phase
levels have been correlated with poorer prognosis III trials.
(Strizzi et al. 2001; Ruotsalainen et al. 2002; Rades A variety of MRI methodologies have been used to
et al. 2010). However, there also are several con- investigate tumor vasculature. These include the use
flicting findings regarding bFGF. These include the of gadolinium (Gd-DTPA) in dynamic contrast
absence of a relationship between bFGF level and enhanced MRI (DCE-MRI), high molecular weight
MVD (Brattstrom et al. 1998; Ueno et al. 2001) and contrast agents to measure vessel permeability and
the lack of correlation between bFGF expression and blood volume, gradient-recalled echo sequences to
survival (Volm et al. 1997). Also, in NSCLC patients, measure a combination of blood oxygenation and
serum bFGF did not differ between clinical stages blood flow (BOLD), and the change in BOLD signal
(Ueno et al. 2001). Finally, one study has reported seen while breathing high oxygen content gases to
that elevated levels of serum bFGF in NSCLC assess vessel maturity (Pathak et al. 2010). Gd-based
patients were related to a better outcome (Brattstrom agents have been used to assess early vascular chan-
et al. 1998). In light of these observations it would ges following anti-angiogeneic treatment. Treatment
appear that the value of bFGF as a surrogate marker with bevacizumab significantly decreased tumor
for tumor angiogenesis in lung cancer remains vascular permeability as measured by gadodiamide
uncertain. (Varallyay et al. 2009). The spatial heterogeneity of
Several other angiogenic molecules, such as matrix response to a VEGF-receptor tyrosine kinase inhibitor
metalloproteinases, epidermal growth factor receptor, was evident in a study using both low and high
angiopoietin-2, thymidine phosphorylase, hepatocyte molecular weight Gd-based contrast agent (Li et al.
growth factor also have been investigated in NSCLC 2005). DCE-MRI has been used in the clinic to
patients. In some of these studies these factors were determine changes following anti-angiogenic treat-
found to be inversely correlated with prognosis (Ab- ment (Batchelor et al. 2007; Jain et al. 2009). How-
delrahim et al. 2010; Dudek et al. 2010; Gordon et al. ever, high molecular weight contrast agents are not
2010). yet readily available clinically, and the BOLD con-
Finally, a range of non-invasive imaging technol- trast method is dependent on the field gradient used,
ogies including ultrasound, positron emission making both comparisons between measurements
tomography (PET), computed tomography (CT) and made on different MR machines and serial measure-
nuclear magnetic resonance imaging (MRI) are ments difficult. DCE-MRI using Gd-DTPA is
available, or under development, that have the becoming increasingly widespread in microcircula-
potential to measure various aspects of tumor vascu- tion research (Hawighorst et al. 1999) and assessment
lature, angiogenesis, and their relation to tumor of changes in microcirculation following treatment
metabolism, proliferation, and growth. intervention (Galbraith et al. 2002, 2003; Jayson et al.
CT can be performed with contrast medium to 2002). Yet this method too has limitations. These are
measure vascular characteristics including blood flow, primarily the consequence of the inherent character-
blood volume, mean fluid transit time, and capillary istics Gd-DTPA which result in the measured
permeability (Miles et al. 2000). Perfusion CT has parameters reflecting a combination of blood flow,
been suggested as a reliable biomarker of tumor vessel permeability and surface area, rather than
angiogenesis in NSCLC (Miles 2003). Perfusion CT being able to discriminate these individual
Angiogenesis and Lung Cancer 23

physiological parameters. Finally, commonly used radiolabeled RGD peptides and the use of PET as
methods lack a directly measured arterial input markers of activated endothelial cell proliferation and
function which affects accuracy and reproducibility of tumor angiogenesis (Schnell et al. 2009; Haubner
the technique (Galbraith et al. 2002). et al. 2010).
The use of PET imaging in oncology is becoming Color Doppler ultrasonography can be used to
widespread, principally using the uptake of 18F measure flow velocity in tumor blood vessels.
labeled fluorodeoxy glucose (FDG) as a measure of Parameters obtained include vascularity index, peak
tumor metabolism. This is proving to be useful in the flow velocity, and flow resistance index. These
assessment of tumor response to therapies, as changes parameters have been used to improve discrimination
in FDG uptake can be detected earlier than traditional between benign and malignant tumors (Strobel et al.
assessment by CT (Kostakoglu and Goldsmith 2003). 2000), to give prognostic information, and to monitor
In NSCLC, PET has advantages over conventional the changes in tumor vascularity after treatment (van
imaging techniques in its ability to discriminate der Woude et al. 1995). Alternatively, ultrasound
mediastinal lymphadenopathy, particularly for techniques using microbubble contrast agents have
assessment of response following radiation therapy also been developed for measurement of blood flow,
(Erdi et al. 2000). and have potential utility in both pre-clinical and
PET methodologies useful for more direct assess- clinical settings (Leong-Poi et al. 2003; Deshpande
ment of tumor vasculature include 15O labeled water et al. 2010). Still, the resolution of ultrasound, and the
for measurement of blood flow, and 11C labeled car- reduced blood velocity in smaller arterioles and cap-
bon monoxide for measurement of blood volume illaries mean that flow in these vessels is not mea-
(Hoekstra et al. 2002). Although the resolution sured by this technique. In addition, bulk tissue
obtained with PET is poorer compared with DCE- movements that produce artifacts can be a problem in
MRI or CT, it has the advantage that absolute blood some organs such as lung (Eriksson et al. 1991).
flow measurements can be obtained. However, the Imaging tumors that are surrounded by aerated lung
very short half life of 15O makes this technique fea- also is technically difficult. Finally, poor accessibility
sible only where a cyclotron is on site. This method to anatomical areas for deep seated tumors, and
has been used for the assessment of response to operator dependence remain challenges for use of
treatment with agents that directly damage tumor these ultrasound methodologies.
vasculature (Anderson et al. 2003).
VG67e, an 124I iodinated monoclonal antibody
which binds to human VEGF-A, was used for 4 Anti-Angiogenic Therapy
assessment of tumor VEGF levels non-invasively
(Collingridge et al. 2002). Similarly, HuMV833, a The complex process of tumor angiogenesis offers
fully human antibody to VEGF-A labeled with 124I, many possible targets for anti-angiogenic strategies.
allows imaging of VEGF distribution in tumors Strategies vary from regulation of angiogenic factor
(Jayson et al. 2002). A variety of different 11C and expression in tumors, to endogenous inhibitors of
18
F-labelled small molecular MMP inhibitors have angiogenesis. There are currently over 3,000 clinical
been developed but, corresponding data from murine trials employing such strategies (
tumor models could not confirm that this class of http://cancertrials.nci.nih.gov/). Based on their bio-
tracers allows the monitoring of tumor-induced logical activities, these strategies can be categorized
angiogenesis (Haubner et al. 2010). Great efforts have into several broad classes. One class of agents spe-
been made to develop radiolabeled RGD peptides for cifically targets angiogenic growth factors. It includes
the non-invasive determination of avb3 expression for tyrosine kinase inhibitors of VEGF/bFGF, as well as
monitoring angiogenesis process. The most studied antibodies or antisense oligonucleotides directed
PET tracer is [18F]Galacto-RGD (Schnell et al. 2009). against pro-angiogenic growth factors or their recep-
Recently, the SPECT tracer [99mTc]NC100692 was tors. A second class of agents includes those designed
introduced by GE healthcare for imaging avb3 to inhibit endothelial cell function, such as thalido-
expression in humans (Bach-Gansmo et al. 2008). The mide and endostatin. A third class consists of matrix
available data indicate significant potential for metalloproteinase inhibitors, compounds that block
24 W. Shi and D. W. Siemann

the degradation of the basement membrane. Agents 15 mg/kg. Because of the serious bleeding events
that target survival factors of neovascular blood sup- during the phase II trial, patients with squamous-cell
ply, such as integrin antagonists comprise yet another carcinoma, and hemoptysis were excluded. Patients
class. with brain metastases were also excluded with the
concern of brain hemorrhage. Response rate was 35%
in the bevacizumab arm and 15% in the control arm.
4.1 Drugs that Block Angiogenic Factors There is a significant improvement in progression-free
survival (6.2 vs. 4.5 m) and overall survival (12.3 vs.
4.1.1 Inhibitors of VEGF and Its Receptors 10.3 m) for the patients treated with bevacizumab.
The central role of VEGF and its receptor system in There were 15 deaths related to bevacizumab com-
tumor angiogenesis has made it a promising target of pared to two associated with chemotherapy (Sandler
anti-angiogenic therapies. Strategies include the use et al. 2006). Nonetheless, the advantages gained
of (i) specific VEGF antibodies to neutralize circu- through the use of bevacizumab far outweigh the risk
lating VEGF, (ii) antisense oligonucleotides or RNA involved.
to disrupt VEGF expression and (iii) VEGF-receptor In the AVAiL (Avastin in Lung Cancer) trial,
antibodies, or receptor associated tyrosine kinase 1,043 patients with advanced nonsquamous cell
inhibitors, to block VEGF signaling (Kim et al. 1993; NSCLC were randomized to six cycles of cisplatin,
Witte et al. 1998; Solorzano et al. 2001; Shi and gemcitabine and bevacizumab or placebo. Bev-
Siemann 2002; Teng et al. 2010). acizumab was administered either 7.5 or 15 mg/kg.
Bevacizumab (Avastin, Genentech Inc., South San The patients continued bevacizumab or placebo as
Francisco, CA) is a monoclonal antibody that targets maintenance until progression (Reck et al. 2009). The
VEGF. It was first approved in metastatic colon bevacizumab group showed significantly improved
cancer when used in combination with chemotherapy progression-free survival, 6.7 and 6.5 months for the
(Hurwitz et al. 2004). It was subsequently approved high-dose and low-dose bevacizumab groups respec-
for use in combination with carboplatin and paclitaxel tively, compared to 6.1 months in placebo group.
for first-line treatment of patients with unresectable There was no significant difference in overall survival
nonsquamous NSCLC (Sandler et al. 2006). New between groups. Subgroup analysis also suggested
studies also provided support for its use with other that the high dose (15 mg/kg) and low dose (7.5 mg/
chemotherapy agents, as well a potential role in the kg) of bevacizumab yielded similar efficacy and
treatment of SCLC (Horn et al. 2009; Patel et al. safety profile. The regimen was overall well tolerated.
2009; Spigel et al. 2009; Jalal et al. 2010). Slightly higher rates of pulmonary hemorrhage of all
Bevacizumab at a dose of 15 mg/kg when com- grades were observed (7% for the low-dose bev-
bined with carboplatin and paclitaxel to previously acizumab group and 9.7% for the high-dose bev-
untreated patients with advanced or recurrent NSCLC acizumab group), compared with 4.9% for
showed promising results in a pivotal phase II trial. It chemotherapy alone group (Reck et al. 2009).
showed a significant increase in time to progression: A recent meta-analysis evaluated the efficacy of
7.4 versus 4.2 months, compared to chemotherapy bevacizumab plus chemotherapy compared to che-
alone (Johnson et al. 2004). However, when tumors of motherapy alone in previously untreated locally
squamous histology, and tumors that were centrally advanced or metastatic NSCLC (Botrel et al. 2011). It
located close to large blood vessels with necrosis or included four trials and 2,200 patients. The response
cavitation were treated with bevacizumab, they rate was higher in patients who received the combi-
showed a tendency to cause bleeding. This resulted in nation of chemotherapy plus bevacizumab 7.5 mg/kg
four fatal episodes of major hemoptysis (Johnson (RR = 0.58; CI95% = 0.46–0.74; p \ 0.00001) and
et al. 2004). Bev 15 mg/kg (RR = 0.53; CI95% = 0.45–0.63;
The role of bevacizumab was fully established in p \ 0.00001) with moderate heterogeneity at dose of
2006 through a phase III trial (E4599) (Sandler et al. 15 mg/kg. The progression-free survival time was
2006). A total of 878 patients with stage IIIB or IV longer in patients who received chemotherapy plus
NSCLC were randomized to paclitaxel and carboplatin bevacizumab 7.5 mg/kg (HR = 0.78, CI95% =
alone or paclitaxel, carboplatin, and bevacizumab at 0.68–0.90; p = 0.0005) and bevacizumab 15 mg/kg
Angiogenesis and Lung Cancer 25

(HR = 0.72, CI95% = 0.65–0.80; p \ 0.00001) with grade 5 events were observed but both were pul-
moderate heterogeneity. Differences in these end monary hemorrhages. Forthcoming trials should
points remained in favor of chemotherapy plus bev- clarify this perspective. The ongoing AVAstin in
acizumab when the analysis was made using the Squamous tumor trial will likely also helps define the
random-effects model. Overall survival was longer in role of bevacizumab and provides answers to the
patients who received CT plus bevacizumab 15 mg/ question of whether radiation therapy before chemo-
kg (HR = 0.89, CI95% = 0.80–1.00; p = 0.04). therapy combined with bevacizumab might minimize
Severe haematologic toxicities (grade [3), neutrope- the risk of bleeding.
nia, and febrile neutropenia were more common Numerous efforts have focused on identifying
among the patients who received bevacizumab. The subgroups of patients that may benefit more from the
study concluded that the combination of chemother- addition of bevacizumab. Biomarker studies accom-
apy plus bevacizumab increased the response rate and panying ECOG 4599 suggest that single nucleotide
progression-free survival of patients with NSCLC. polymorphism in VEGF, EGF, intercellular adhesion
With respect to overall survival the benefits of bev- molecule-1, and WNK lysine deficient protein kinase
acizumab remains uncertain (Botrel et al. 2011). 1 may predict response (Zhang et al. 2009). Hyper-
Currently, bevacizumab is being evaluated for tension, one side effect of bevacizumab treatment has
combination with other chemotherapeutic agents, also been suggested to be a biomarker of clinical
such as docetaxel, oxaliplatin, and pemtrexed. The benefit of bevacizumab from several studies
combination of bevacizumab with carboplatin and (Schneider et al. 2008a; Rini et al. 2010; Osterlund
pemetrexed, followed by maintenance pemerexed and et al. 2011). However, contrary to these observation, a
bevacizumab showed impressive overall response rate retrospective analysis of six trials in colorectal, breast,
of 55%, a progression-free and median survival time and renal cell carcinoma showed that treatment
of 7.8 and 14.1 months, respectively (Patel et al. induced hypertension was predictive of overall sur-
2009). It becomes one primary option for patients vival and progression-free survival in only one study
with the characteristics described in the trial. (Hurwitz et al. 2010). The predictive value of bev-
Bevacizumab also been evaluated in the mainte- acizumab-induced hypertension might not extend to
nance setting in the management of NSCLC. The all cancers or all treatment regimens. The measure-
ATLAS phase III trial compared bevacizumab with or ment of concentrations of circulating protein is an
without erlotinib after completion of a first-line che- attractive biomarker strategy, as blood is easily
motherapy regimen that included bevacizumab accessible. However, concentrations of circulating
(Miller VAOCP et al. 2009). The trial was stopped VEGF before treatment were not correlated with
early because progression-free survival was efficacy of bevacizumab in an analysis of four phase
4.8 months for the combined arm and 3.7 months for III trials (Bernaards et al. 2010). Still, VEGF levels
the bevacizumab alone arm (p = 0.0012). Overall are dynamic, and changes related to pretreatment
survival was not significantly different. values might have predictive value (Loupakis et al.
Currently, bevacizumab is contraindicated for 2007; Zahiragic et al. 2007; Lu et al. 2008; Willett
patients with brain lesions or squamous carcinomas et al. 2009). Changes in concentrations of numerous
owing to the risk of cerebrovascular bleeding and other proteins putatively related to angiogenesis also
hemoptysis. However, the prospective PASSPORT have been documented after the start of bevacizumab
trial raised another perspective concerning the safety treatment (Wedam et al. 2006; Dowlati et al. 2008;
of bevacizumab in the setting of brain metastases Willett et al. 2009; Jubb and Harris 2010). Currently,
(Socinski et al. 2009). In this study, treatment of na only circulating intercellular adhesion molecule one
patients with previously treated brain metastases has shown any predictive value in terms of survival
received bevacizumab with platinum-based doublet benefit with bevacizumab in a phase III trial (Dowlati
therapy or erlotinib at physician’s discretion. Second- et al. 2008; Jubb and Harris 2010). Polymorphisms of
line patients received either bevacizumab with single- components of the VEGF pathway also have been
agent chemotherapy or erlotinib at physician’s dis- proposed to predict benefit from bevacizumab treat-
cretion as well. For 106 patients, no grade C2 central ment (Schneider et al. 2008b; Schultheis et al. 2008).
nervous system hemorrhages were reported; two However, these findings have not been validated and
26 W. Shi and D. W. Siemann

the prognostic importance of these polymorphisms in and 12 month survival rates were 54 and 29%,
patients treated undergoing bevacizumab therapy has respectively. Common grade 3/4 toxicities included
not been established. dyspnea (21%), hypertension (23%), and proteinuria
Bevacizumab has also been evaluated in the (10%). Two cases of grade 5 hemoptysis were
management of SCLC. For example, one ECOG study reported, as well as one case each of tracheoesopha-
added bevacizumab to cisplatin and etoposide for up geal fistula, decreased cardiac ejection fraction,
to four cycles and continued bevacizumab as main- cerebral ischemia, and reversible posterior leukoen-
tenance for up to 1 year in patients with extensive- cephalopathy. Aflibercept has minor single agent
stage SCLC (Horn et al. 2009). The study showed a activity in heavily pretreated lung adenocarcinoma,
median progression-free survival of 4.7 months, but appears to be well tolerated with no unexpected
median overall survival of 10.9 months, and 1 year toxicities. Phase III trials in this setting are ongoing.
overall survival of 38.1%. This outcome compares Other efforts are exploring the role of aflibercept in
favorably with historical data established with etop- lung cancer in combination with platinum-based
side-cisplatin alone. In another multicenter phase II doublets and single-agent docetaxel (Riely and Miller
trial, patients with extensive SCLC received irino- 2007).
tecan, carboplatin, and bevacizumab. Patients who Ramucirumab, (IMC-1121B, ImClone systems
had no progression also received maintenance bev- Inc) is a fully humanized monoclonal antibody that
acizumab (Spigel et al. 2009). The objective respon- blocks the binding of the VEGF ligand to the extra-
sive rate was 84%, median time to progression was cellular domain of VEGFR-2. A phase I trial in
9.13 months, median overall survival was patients with advanced solid malignancies has shown
12.1 months, and 1 and 2 year overall survivals were it to be well tolerated with favorable response rates
51 and 14%, respectively. This result compares (Spratlin et al. 2010). It is currently in Phase II studies
favorably with larger randomized trials using che- in colorectal, prostate, liver, non-small-cell lung and
motherapy alone. Another recent trial from the Hoo- ovarian cancers, as well as melanoma and recurrent
sier Oncology Group included patients with relapsed glioblastoma multiforme as well as three phase III
chemosensitive SCLC (Jalal et al. 2010). Patients studies in breast cancer (NCT00703326), gastric
receiving paclitaxel and bevacizumab had a median cancer or gastroesophageal junction adenocarcinoma
survival time of 30 weeks and the addition of bev- (NCT00917384), and hepatocellular carcinoma
acizumab to paclitaxel did not improve outcomes in (NCT01140347). Three additional phase III studies of
relapsed chemosensitive SCLC. Thus the role of ramucirumab with or without paclitaxel in metastatic
bevacizumab remains not fully defined in SCLC. gastric adenocarcinoma (NCT01170663), in second-
Aflibercept (VEGF-trap/AVE0005, Regeneron line metastatic colorectal cancer (NCT01183780), and
Pharmaceuticals, Tarrytown, NY) is a fully human in second-line non-small-cell lung cancer
soluble fusion protein that binds circulating VEGF-A (NCT01168973) are planned.
and placental growth factor, preventing binding to the IMC-18F1 (ImClone Systems, Inc) is a humanized
cell surface membrane receptors (Teng et al. 2010). It monoclonal antibody against VEGFR-1 that has
is engineered by combining domains from VEGFR-1 demonstrated antitumor activity in preclinical studies
and VEGFR-2 with the Fc domain of human IgG (Schwartz et al. 2010). A phase I study of patients
(Riely and Miller 2007). A phase I trial of aflibercept with advanced solid tumors who no longer responded
showed dose-limiting toxicities of rectal ulceration to standard therapy just closed, and the results are
and proteinuria at a 7 mg/kg dose given intravenously pending. Three phase II studies are open in colorectal,
every 2 weeks, and 4 mg/kg was advanced as the breast, and bladder, urethra, ureter, or renal pelvis
phase II dose (Lockhart et al. 2010). In a multicenter carcinoma.
phase II trial evaluating the efficacy and safety of An alternative approach to interrupt VEGF activity
intravenous aflibercept in patients with platinum- and that has received a great deal of attention is the use of
erlotinib-resistant lung adenocarcinoma (Leighl et al. small molecule compounds to inhibit VEGF-receptor
2010) 98 patients were enrolled; 89 were evaluable associated tyrosine kinases. Most of these agents bind
for response. Median progression-free survival was to the ATP-binding site of the receptor, thus inhibit-
2.7 months, and overall survival was 6.2 months. Six ing the activation of the receptor and subsequent
Angiogenesis and Lung Cancer 27

Table 2 Tyrosine kinase inhibitors and their targets


TKI VEGFR1 VEGFR2 VEGFR3 PDGFR EGFR KIT Other
Vandetanib + + + RET
Cediranib + + + + +
Sorafenib + + + + + RAF, FLT3
Sunitinib + + + + + FLT3, RET
Axitinib + + +
Vatalanib + + + + +
Motesanib + + + + + RET
Pazopanib + + + + +
BIBF1120 + + FGFR

downstream signaling. In addition to inhibiting 0.70–0.90; p \ 0.0001); median PFS was 4.0 months
VEGFR2 associated signaling, many such inhibitors in the vandetanib group versus 3.2 months in the
target other tyrosine kinases. Simultaneous targeting placebo group. The trial also showed that the addition
of several kinases offers a theoretical advantage over of vandetanib to docetaxel provided a significant
single kinase inhibitors because most cancers have improvement in PFS in patients with advanced
complex and often redundant signaling pathways. NSCLC after progression following first-line therapy
However, a potential disadvantage is the potential for (Herbst et al. 2010). The ZEAL trial enrolled 534
toxicity resulting from off-target kinase inhibition; a patients, who were randomized to receive vandetanib
possibility that may have particular relevance when with pemetrexed or placebo with pemetrexed. The
these agents are combined with chemotherapy. results showed that there was no significant difference
Table 2 provides a listing of the stages of clinical in PFS or overall survival between the treatment
development of various anti-angiogenic tyrosine arms. Statistically significant improvements in
kinase inhibitors and their targets. objective response rate (19 vs. 8%; p \ 0.001) and
Vandetanib (Zactima, AZD6474, AstraZeneca time to deterioration of symptoms (HR, 0.71;
Pharmaceuticals, Wilminton, DE) is an oral anilino- p = 0.0052; median, 18.1 weeks for vandetanib and
quinazoline that inhibits VEGFR1, VEGFR2, VEG- 12.1 weeks for placebo) were observed in patients
FR3, RET, and EGFR (Ciardiello et al. 2003). Phase I receiving vandetanib. Adding vandetanib to pemetr-
studies identified a maximum tolerated daily dose of exed also increased the incidence of some adverse
300 mg. Hypertension and prolongation of QTc were events, including rash, diarrhea, and hypertension,
the most common adverse effects (Holden et al. while showing a reduced incidence of nausea, vom-
2005). In a randomized phase II trial versus gefitinib, iting, anemia, fatigue, and asthenia with no reduction
progression-free survival was 11 and 8.1 weeks for in the dose intensity of pemetrexed. The vandetanib
vandetanib and gefitinib, respectively (Natale et al. combination showed a significantly higher objective
2009). In another phase II trial, two doses of vande- response rate and a significant delay in the time to
tanib were tested with docetaxel compared with worsening of lung cancer symptoms versus the pla-
docetaxel alone in NSCLC patients previously treated cebo arm as well as an acceptable safety profile in this
with platinum-based chemotherapy. The progression- patient population. However, this study did not meet
free survival favored the two arms with vandetanib the primary end point of statistically significant PFS
(Heymach et al. 2008). These results led to four phase prolongation with vandetanib plus pemetrexed versus
III trials, ZEST, ZEAL, ZEPHYR, and ZODAIC. The placebo plus pemetrexed (de Boer et al. 2011). The
ZODAIC trial enrolled 1,391 patients, who were ZEST trial included 1,240 patients, who were ran-
randomized to vandetanib with docetaxel or placebo domized to vandetanib 150 or 300 mg/d, or erlotinib.
with docetaxel. Vandetanib plus docetaxel led to a There was no significant improvement in PFS for
significant improvement in PFS versus placebo plus patients treated with vandetanib versus erlotinib
docetaxel (hazard ratio [HR] 0.79, 97.58% CI (hazard ratio [HR], 0.98; 95.22% CI, 0.87–1.10;
28 W. Shi and D. W. Siemann

p = 0.721); median PFS was 2.6 months for vande- NSCLC after failure with two prior regimens of
tanib and 2.0 months for erlotinib. There was also no chemotherapy. After a 2 month lead in period during
significant difference for the secondary end points of which all patients received active drug, those with
overall survival (HR, 1.01; p = 0.830), objective stable disease were randomized to sorafenib or pla-
response rate (both 12%), and time to deterioration of cebo. The median progression-free survival was 3.6
symptoms for pain (HR, 0.92; p = 0.289), dyspnea and 2 months in the sorafenib and placebo arms,
(HR, 1.07; p = 0.407), and cough (HR, 0.94; respectively (Clement-Duchene and Wakelee 2010).
p = 0.455). Both agents showed equivalent PFS and A phase III trial of sorafenib versus placebo is
overall survival in a preplanned non-inferiority anal- ongoing.
ysis. The trial concluded that in patients with previ- Sunitinib (Sutent, SU11248, Pfizer Inc, New York)
ously treated advanced NSCLC, vandetanib showed is an inhibitor of VEGFR, PDGFR, KIT, FLT3, RET,
antitumor activity but did not demonstrate an efficacy and CSF-1R. It is approved for the treatment of
advantage compared to erlotinib. There was a higher advanced renal cell carcinoma and imatinib-resistant
incidence of some adverse effects with vandetanib gastrointestinal stromal tumors (Heng and Koll-
(Natale et al. 2011). The ZEPHYR trial randomized mannsberger 2010). In a phase II trial, 63 patients
patients to vandetanib or placebo. The results reported with NSCLC received daily 50 mg doses of sunitinib
in abstract form showed that vandetanib improved initially for 4 weeks then followed by 2 weeks of
progression-free survival by nearly 40%. However, treatment in a 6 week cycle. The overall response rate
there was no improvement in overall survival and was 11.1%. The median progression-free survival and
since this was the primary study endpoint, it was overall survival were 12 and 23.4 weeks, respectively
considered a negative trial. (Socinski et al. 2008). In another phase II trial patients
Cediranib (Recentin, AZD2171, AstraZeneca with advanced NSCLC received continuous daily
Pharmaceuticals, Wilminton, DE) is a potent VEGFR2 dosing of sunitinib (37.5 mg). Median PFS was
inhibitor that also inhibits VEGFR1, VEGFR3, and 11.9 weeks (95% CI 8.6, 14.1) and median OS was
PDGFR (Wedge et al. 2005). As a single agent, it was 37.1 weeks (95% CI 31.1, 69.7). The 1 year survival
well tolerated at doses up to 45 mg given daily (Laurie probability was 38.4% (95% CI 24.2, 52.5). Treat-
et al. 2008). In a phase II chemotherapy combination ment was generally well tolerated (Novello et al.
trial in patients with previously untreated advanced 2009). A phase III trial investigating sunitinib and
stage NSCLC, the response rate was high (38%). erlotinib combination is ongoing.
However, increased toxicities were reported when Axitinib (AG-013736, Pfizer Inc. New York) is a
daily dose of 30 mg were given (Goss et al. 2010). As a tyrosine kinase inhibitor with activity against VEG-
result the BR.29 trial was recently opened using a daily FR, PDGFR, and c-kit (Hu-Lowe et al. 2008; Kelly
dose of 20 mg (http://australiancancertrials.gov.au). and Rixe 2010). In a phase I study, the maximum
Sorafenib (Nexavar, BAY 43-9006, Bayer Phar- tolerated dose was 5 mg when given twice daily. The
maceuticals Corporation, West Haven, CT) is a raf main adverse effects were hypertension, seizure,
and VEGFR inhibitor with activity against PDGFR abnormal liver functions, and mesenteric vein
and KIT. It has proven activities in renal cell carci- thrombosis with pancreatitis (Rugo et al. 2005). In a
noma and hepatocellular carcinoma (Escudier et al. phase II study that evaluated the efficacy of axitinib as
2007; Rimassa and Santoro 2009). In the phase III a single agent in NSCLC, the response rate was 41%,
ESCAPE trial, which randomize patients to carbo- progression-free survival was 4.9 months, and median
platin and paclitaxel, with and without sorafenib in overall survival was 14.8 months (Schiller et al.
first-line treatment for advanced NSCLC, the primary 2009). Phase II evaluations of axitinib plus paclitaxel
endpoint of overall survival was not met. The study and carboplatin, and axitinib plus cisplatin and
was terminated early as a result of the detrimental pemetrexed as well as a phase III trial evaluating
effect of sorafenib on patients with squamous-cell axitinib as a single agent in advanced NSCLC are
carcinoma and lack of efficacy in nonsquamous car- ongoing. Vatalanib (ptk787, zk-222584, Novartis/
cinoma patients (Scagliotti et al. 2010). A large phase Schering AG, Berlin, Germany) is a VEGFR,
II trial, E2501, enrolled more than 300 patients and PDGFR, and KIT inhibitor (Scott et al. 2007). It is
compared sorafenib with placebo in patients with currently being studied in phase II/III trials. In a phase
Angiogenesis and Lung Cancer 29

II study of vatalanib monotherapy in previously angiokinase inhibitor BIBF 1120 in patients with
treated NSCLC, this agent showed moderate efficacy. relapsed advanced non-small-cell lung cancer. The
The response rate was 10% and overall survival was median PFS was 6.9 weeks, with no significant dif-
7 months (Gauler et al. 2007). A phase I/II trial of ference between treatment arms. Median overall sur-
vatalanib and pemetrexed with or without cisplatin for vival (OS) was 21.9 weeks. Continuous treatment
lung cancer is ongoing. with BIBF 1120 was well tolerated, with no difference
Motesanib (AMG 706. Amgen, Thousand Oaks, in efficacy between treatment arms. PFS and objective
CA) is an orally active multikinase inhibitor (Polve- response with single-agent treatment in advanced
rino et al. 2006). Phase I studies in solid tumors disease warrants further exploration (Reck et al.
showed a maximum tolerated daily dose of 125 mg 2011). Phase III placebo control trials of BIBF 1120
(Rosen et al. 2007). A phase II trial randomized in combination with docetaxel or pemetrexed are in
patients with advanced NSCLC to motesanib or progress.
bevacizumab in combination with paclitaxel and Finally, ribozyme constructs that target VEGF-
carboplatin. The efficacy of daily motesanib or bev- receptor mRNA are also under development. Pre-
acizumab plus carboplatin and paclitaxel was esti- clinical studies with these constructs induced inhibi-
mated to be comparable. Toxicity was higher but tion of growth in both primary and metastatic Lewis
manageable in both motesanib arms. Efficacy and lung carcinoma (Oshika et al. 2000; Pavco et al. 2000;
tolerability of daily 125 mg doses of motesanib plus Fabbro and Manley 2001). A phase I trial of anti-Flt-1
carboplatin and paclitaxel in advanced nonsquamous ribozymes was carried out in patients with advanced
NSCLC were investigated in a phase III study. This cancer. Minor clinical responses were observed with
trial was closed due to a higher early mortality and a 14 of 20 patients maintaining stable disease for 1–
higher rate of hemoptysis in patients with squamous 6 months (Fabbro and Manley 2001).
histology but was reopened with exclusion of this
patient population. 4.1.2 Non-Specific Agents
Pazopanib (GW786034, GlaxoSmithKline, Phi- Thalidomide (Celgene, Summit, NJ) has been shown
ladephia) is a VEGF, PDGFR, and KIT inhibitor to inhibit angiogenesis, though the mechanism of
currently in phase III development for advanced renal action is poorly understood. It may be mediated
cell carcinoma and phase II development for through inhibition of TNF-a, VEGF, and bFGF
NSCLC.(Altorki et al. 2010; Sternberg et al. 2010). In expression by tumor cells, cell surface receptors
a phase II trial proof-of-concept study, it examined inhibition, and/or effects on the immune system
safety and efficacy of short-term, preoperative paz- (D’Amato et al. 1994; Li et al. 2003). Thalidomide
opanib monotherapy in patients with operable stage I/ has shown limited efficacy in a phase II NSCLC trial
II NSCLC. Patients scheduled for resection received in combination with first-line carboplatin/irinotecan
oral pazopanib 800 mg/d for 2–6 weeks preopera- (Miller et al. 2006). In another phase II trial, thalid-
tively. Short-duration pazopanib was generally well omide was used with irinotecan and gemcitabine in
tolerated and demonstrated single-agent activity in chemonaive patients with advanced NSCLC. The
patients with early stage NSCLC. Several target genes median time to disease progression was 4 months.
were dysregulated after pazopanib treatment, vali- The 1 and 2 year survival rates were 36 and 27%,
dating target-specific response and indicating a per- respectively (Jazieh et al. 2009). Thalidomide was
sistent pazopanib effect on lung cancer tissue. Further also evaluated in the setting of neoadjuvant therapy
clinical evaluation of pazopanib in NSCLC is planned with carboplatin, gemcitabine in stage IIB and III
(Altorki et al. 2010). NSCLC. Response rates were 70% and overall sur-
BIBF1120 (Vargatef, Boehringer-Ingelheim, In- vival was 3.6 years (Dudek et al. 2009). Unfortu-
gelheim, Germany) is an oral indolinone derivative nately, in a large randomized double-blind placebo-
that inhibits VEGFR2, FGFR, and PDGFR. Phase I controlled trial of thalidomide in combination with
trial investigations established a 200 mg twice daily gemcitabine and carboplatin in advanced NSCLC,
dose for subsequent phase II evaluation (Mross et al. thalidomide in combination with chemotherapy did
2010). A phase II double-blind study investigated not improve survival overall, but increased the risk of
efficacy and safety of two doses of the triple thrombotic events (Lee et al. 2009). Thalidomide was
30 W. Shi and D. W. Siemann

also evaluated in the maintenance therapy for exten- failed to improve survival (Mutter et al. 2009). A phase
sive-stage SCLC after response to chemotherapy. II study of celecoxib and docetaxel in NSCLC patients
Thalidomide 200 mg daily is well tolerated when with progression after platinum-based therapy also
given as maintenance therapy for ES-SCLC after failed to improve the response rate and survival com-
induction chemotherapy (Dowlati et al. 2007). pared with docetaxel alone (Schneider et al. 2008).
Lenalidomide (Revlimid, CC-5013. Celgene, Apricoxib, another COX2 inhibitor, was evaluated in a
Summit, NJ) is a thalidomide analogue that is a phase I trial in combination with erlotinib in advanced
licensed for the treatment of multiple myeloma and NSCLC. Apricoxib plus erlotinib was well tolerated
myelodysplastic syndrome (Galustian and Dalgleish and yielded a 60% disease control rate. A phase II trial
2009). A phase I study of lenalidomide in solid is currently investigating 400 mg/day dose of apric-
tumors led to a recommended dosing protocol of oxib plus 150 mg/day erlotinib in patients selected on
25 mg/day for 4 weeks followed by a 2 week rest the basis of in urinary PGE-M changes (Reckamp et al.
period for lenalidomide use in patients with solid 2011).
tumors (Miller et al. 2007). Lenalidomide was also
evaluated in a phase I trial in combination with
docetaxel and carboplatin in patients with advanced 4.2 Drugs that Inhibit Endothelial Cell
solid tumors. The results showed that in combination Function
with docetaxel 60 mg/m2 and carboplatin AUC 6 on
Day 1, lenalidomide 5 mg orally daily on days 1–14 Endostatin, a 20 kDa C-terminal proteolytic fragment
of a 21-day cycle was the maximum tolerated dose of collagen XVIII, has been identified as a potent
without the use of prophylactic growth factors (Kal- endogenous inhibitor of angiogenesis. In murine
madi et al. 2007). A phase II trial (NCT00179686) in models, the growth of Lewis lung tumors was mark-
NSCLC was completed and results are pending. edly suppressed by systemic endostatin therapy. At a
Pomalidomide (Actimid, CC-4047, Celgene, NJ), a dose of 20 mg/kg once daily, there was almost com-
third generation thalidomide analogue (Lacy and plete regression of established primary tumors
Tefferi 2011), is being evaluated in a phase I/II study (O’Reilly et al. 1997). However, in patients, no clin-
as maintenance therapy in patients with extensive- ical responses were observed (Thomas et al. 2003). A
stage SCLC (NCT00537511). few patients did demonstrate changes in their
Cyclooxygenase 2 (COX2), an enzyme that is dynamic CT scans suggestive of a decline in micro-
involved in prostaglandin synthesis, is frequently up- vessel density, but overall no consistent effect of
regulated in NSCLC, and may be a marker of worse endostatin on tumor vasculature was seen. Other
prognosis (Dannenberg et al. 2001). It also may pro- studies have noted measurable effects of endostatin on
mote angiogenesis, prevent apoptosis, and induce tumor blood flow and metabolism and the induction
resistance to radiation therapy. Inhibitors of COX2 of tumor and endothelial cell apoptosis but again
have been widely used for inflammatory conditions, these occurred in the absence of demonstrable anti-
and their anti-cancer activity has only recently begun to tumor effects (Herbst et al. 2002). Endostar is a novel
be explored. A phase II study in combination with recombinant human endostatin expressed and purified
preoperative paclitaxel and carboplatin in patients with in Escherichia coli with an additional nine-amino acid
Stage I–IIIA NSCLC (Altorki et al. 2003) reported sequence and forming another his-tag structure (Ling
encouraging response rates, but definitive demonstra- et al. 2007). It was evaluated in a phase II study of
tion of the potential benefit of such combinations cisplatin/etoposide for extensive-stage small-cell lung
awaits randomized trials. Another phase II trial eval- cancer. The addition of rh-endostain to cisplatin and
uated celecoxib in combination with paclitaxel, car- etoposide in patients with ED-SCLC results in
boplatin, and radiotherapy in patients with inoperable slightly improved PFS and OS relative to historical
stage IIIA/B non-small-cell lung cancer, but this trial controls who received this chemotherapy regimen
was terminated because it did not meet the predeter- alone. This regimen appears to be well tolerated
mined goal of 80% overall response rate. In unselected (Zhou et al. 2011).
patients, the addition of celecoxib to concurrent che- TNP-470 (Takeda Chemical Industries Ltd, Osaka,
moradiotherapy with inoperable stage IIIA/B NSCLC Japan), a synthetic analog of fumagillin, is an
Angiogenesis and Lung Cancer 31

angiogenesis inhibitor that blocks the growth of new showed no significant difference in survival (Shep-
blood vessels by inhibiting methionine aminopepti- herd et al. 2002). Similarly, Prinomastat (AG3340), a
dase, an enzyme critically important for endothelial more selective MMP inhibitor with activity against
cell proliferation (Sin et al. 1997). In the clinic, partial MMP-2, 3, 9, and 14 failed to demonstrate efficacy in
responses were observed in 6 out of 16 NSCLC stage IIB/IV NSCLC patients (Bissett et al. 2005). A
patients treated with TNP470 (Herbst et al. 2002) phase I study with CGS 27023A (MMI270), a MMP
suggesting that further evaluation of TNP-470, par- inhibitor with activity against MMP-1, 2, 3, 9, and 13,
ticularly in combination with chemotherapy, may be that was carried out in patients with solid tumors,
warranted. including lung cancer patients (Levitt et al. 2001),
also resulted in no positive tumor responses. Finally,
when BAY12-9566, an inhibitor of MMP-2 and 9 was
4.3 Drugs that Block Breakdown evaluated in SCLC and stage III NSCLC patients,
of Extracellular Matrix both trials were closed before reaching their accrual
goal because the results showed a detrimental effect
To form new blood vessels, endothelial cells of on patient survival (Hidalgo and Eckhardt 2001).
existing blood vessels must degrade the underlying Studies with two other MMP inhibitors are
basement membrane and invade into the stroma of the ongoing. Neovastat (AE-941) is a naturally occurring
neighboring tissue. The processes of endothelial cell MMP inhibitor derived from shark cartilage extract
invasion and migration require the cooperative that has shown antitumor/antimetastatic properties in
activity of plasminogen activators and matrix animal models and few side effects in more than 800
metalloproteinases (MMPs). The MMPs are a family patients (Gingras et al. 2003). A phase III random-
of structurally related zinc-dependent endopeptideas- ized study of induction platinum-based chemother-
es collectively capable of degrading extracellular apy and radiotherapy with or without neovastat in
matrix. Their activities are controlled at different patients with unresectable stage IIIA or IIIB NSCLC
levels (Liekens et al. 2001; Gialeli et al. 2011): (i) has now been completed. The addition of neovastat
their expression is up-regulated by angiogenic growth to chemo-radiotherapy did not improve overall sur-
factors, (ii) they need to be activated proteolytically, vival in patients with unresectable stage III NSCLC.
and (iii) their activities are negatively impacted by This study does not support the use of shark carti-
their inhibitors (tissue inhibitors of metalloproteinases lage-derived products as therapy for lung cancer (Lu
(TIMPs)). Ultimately, an imbalance between MMPs et al. 2010). Another agent, BMS-275291, inhibits a
and TIMPs is responsible for an invasive phenotype broad range of MMPs known to be associated with
(Gialeli et al. 2011). In light of such rationale, the the growth and spread of tumors (Poulaki 2002). It is
inhibition of MMPs has been extensively studied as currently in phase II/III trials, as an adjunct to
an approach to inhibit the growth and invasion of standard chemotherapy, in advanced or metastatic
neoplastic cells (Vihinen and Kahari 2002). Unfor- NSCLC patients. In a randomized phase III NCI-
tunately, clinical outcomes with these agents have Canada-clinical trials group sponsored study (BR.18)
been large and very disappointing. BMS-275291 in combination with paclitaxel and
Marimastat was the first orally administrated syn- carboplatin in advanced NSCLC increased toxicity
thetic MMP inhibitor and was the first to be evaluated and but failed to improve survival (Leighl et al.
in SCLC. It inhibits the activity of MMP-1, 2, 3, 7, 2005).
and 9. The principle toxicity of marimastat observed
in several phase I–II clinical studies was the appear-
ance of a dose-limiting inflammatory polyarthritis that 4.4 Drugs that Target Survival Factors
consisted of joint stiffness and pain (Steward 1999). of Neovessels
Marimastat was tested in two phase III SCLC studies
in which patients were treated with chemotherapy A number of factors influence endothelial cell sur-
with or without thoracic radiotherapy. After com- vival with VEGF being perhaps the most notable.
pleting the cytotoxic therapy, patients were random- Indeed it is now recognized that anti-VEGF thera-
ized to receive placebo or marimastat. The results peutic approaches, in addition to their other actions,
32 W. Shi and D. W. Siemann

may directly affect endothelial cell survival (Gerber example, endothelial cell specific promoter elements
et al. 1998; Caron et al. 2009). and vectors with restricted cellular tropisms have
Another approach that also aims to affect endo- been examined (Trepel et al. 2000). The strategy of
thelial cell survival targets integrins. Integrins are linking antibodies or peptides that recognize tumor-
heterodimeric transmembrane proteins consisting of a associated vasculature to toxins or pro-coagulant/pro-
and b subunits with large ectodomains and short apoptotic effector molecules that can induce endo-
cytoplasmic tails. They control cell motility, differ- thelial cell damage also has been explored. The utility
entiation, and proliferation via interactions with of such ligand-directed targeting is supported by
extracellular matrix molecules. Integrins avb3 and recent in situ studies in preclinical tumor models that
avb5 are up-regulated on proliferating endothelial demonstrated not only the localization of the thera-
cells in angiogenic blood vessels (Brooks et al. 1994). peutic moiety to tumor vessels but also the induction
The avb3 integrin, an adhesion receptor for extracel- of thrombi formation and the selective destruction of
lular matrix components with an exposed RGD vasculature (Nilsson et al. 2001; Veenendaal et al.
sequence, is an attractive target for anti-angiogenic 2002).
therapy because it is almost exclusively present on the In the category of small molecule drugs, two
cell surface of activated endothelial cells. It is con- classes of agents that selectively disrupt the tumor
sidered a survival factor for angiogenic vessels vessel network have entered clinical trials. The first
(Eliceiri and Cheresh 1999). Antibodies against avb3 includes flavone acetic acid (FAA) and its potent
have been shown to inhibit adhesion-dependent signal analog 5,6-dimethylxanthenone-4-acetic acid
transduction by angiogenic factors, leading to apop- (DMXAA, vadimezan, ASA404) (Baguley 2003;
tosis of activated endothelial cells. Consequently, Baguley and McKeage 2010). The mechanism of
these agents block endothelial tube formation and action of these agents appears to be largely indirect,
angiogenesis in tumors (Brooks et al. 1994). Cilen- through the induction of cytokines, particularly TNF-
gitide (EMD 121974, Merck, Darmstadt, Germany), a a (Philpott et al. 1997; Ching et al. 2004; Baguley and
cyclic RGD pentapeptide, is an inhibitor of v3 and v5 Siemann 2010), although the ability of ASA404 to
integrin receptors. No efficacy data are currently induce targeted pro-inflammatory response within
available in lung cancer. tumor tissue may also critically contribute to its
action (Philpott et al. 1997; Ching et al. 2004;
Baguley and Siemann 2010). Vadimezan was evalu-
5 Vascular Disrupting Therapies ated in a phase II study combined with carboplatin
and paclitaxel in previously untreated NSCLC. The
An alternative to targeting tumor blood vessels on the best overall tumor response was partial response,
basis of interfering with the process of tumor cell which was seen in 37.9% of patients by independent
induced new vessel formation (i.e., anti-angiogenic assessment and in 46.7% by investigator assessment.
therapies) is to develop agents that specifically com- Median time to tumor progression was 5.5 months by
promise the function of existing vasculature in solid investigator assessment and median survival was
tumors. Such approaches aim to cause direct damage 14.9 months (McKeage et al. 2009). The phase II
to the established tumor endothelium and thus lead to results led to its advancement to two randomized,
extensive secondary neoplastic cell death (Denekamp double-blind, placebo-controlled phase III trials in
1990; Siemann and Shi 2003). These vascular dis- advanced NSCLC; the first (ATTRACT-1) as first-
rupting agents and their therapeutic application may line therapy and the second (ATTRACT-2) as second-
be broadly divided into two categories: biological line therapy (McKeage et al. 2009). Both combina-
agents and small molecule drugs. tions employed vadimezan at a dose of 1,800 mg/m2
Biological approaches include targeted gene ther- (calculated as the free base) for the second treatment
apy, antibodies to neovascular antigens, and fusion arm and were administered every 3 weeks for up to
proteins targeting specific endothelial cell receptors. six cycles. The ATTRACT-1 study utilized paclitaxel
Although investigations utilizing these approaches (200 mg/m2) and carboplatin (AUC 6 mg/ml/min) as
have, to date, been confined to preclinical investiga- standard therapy. There were no safety concerns and
tions, encouraging results have been reported. For no unexpected adverse effects when compared with
Angiogenesis and Lung Cancer 33

the phase II data, but the trial was halted when interim paclitaxel, bevacizumab, and fosbretabulin in patients
data analysis failed to show a survival advantage ( with chemotherapy na lung cancer.
http://www.antisoma.com). The ATTRACT-2 trial,
which utilizes docetaxel (75 mg/m2) as standard
therapy in patients with Stage IIIb/IV disease remains 6 Conclusions
ongoing.
The second class includes a group of tubulin- Angiogenesis plays a critical role in the progression
binding agents, most notably combrestastatin A4 and prognosis of lung cancer. Although still in early
disodium phosphate (Fosbretabulin, CA4DP) and the stages of development, therapeutic strategies directed
phosphate prodrug of N-acety-colchinol (ZD6126). against the tumor blood vessel network represents a
The principal mechanism of action of this class of promising advance in the management of lung cancer
drugs is believed to be the selective disruption of the patients. The recent demonstration of improvement in
cytoskeleton of proliferating endothelial cells that survival in colorectal cancer with bevacizumab
leads to thrombus formation and a secondary ische- treatment is the first clinical validation of anti-
mic tumor cell death (Galbraith et al. 2001; Kanthou angiogenic therapy, providing hope that similar ben-
and Tozer 2002; Siemann 2011). Fosbretabulin was efits may be seen in other tumor types including lung
evaluated in a phase I trail with carboplatin and cancer. Ultimately, such endeavors are likely to
paclitaxel in patients with advanced cancer. Dose- incorporate both anti-angiogenic and vascular dis-
limiting toxicity of grade 3 hypertension or grade 3 rupting strategies in combination with conventional
ataxia was seen in two patients at 72 mg/m2. anti-cancer measurements.
Responses were seen in 22% patients with ovarian,
oesophageal, small-cell lung cancer, and melanoma
(Rustin et al. 2010). Phase II trial in NSCLC are
ongoing. ZD6126 is currently under phase II trial.
References
Other vascular disrupting agents, including
AVE8062, NPI-2358, ABT-751, TZT-1027, CYT997, Abdelrahim M, Konduri S et al (2010) Angiogenesis: an update
and potential drug approaches (review). Int J Oncol
Dolastatin 10, MPC-6827, OXi4503, EPC2407, MN- 36(1):5–18
029, and BNC105 are in early clinical trial develop- Altorki NK, Keresztes RS et al (2003) Celecoxib, a selective
ment (Siemann 2011). cyclo-oxygenase-2 inhibitor, enhances the response to
Optimal treatment strategies with agents that preoperative paclitaxel and carboplatin in early stage non-
small-cell lung cancer. J Clin Oncol 21(14):2645–2650
damage the existing tumor vessel network will ulti- Altorki N, Lane ME et al (2010) Phase II proof-of-concept
mately likely combine such therapeutics with con- study of pazopanib monotherapy in treatment-naive patients
ventional therapies including radiotherapy and with stage I/II resectable non-small-cell lung cancer. J Clin
chemotherapy for maximum treatment effect (Sie- Oncol 28(19):3131–3137
Anagnostou VK, Tiniakos DG et al (2011) Multiplexed analysis
mann and Horsman 2002; Siemann and Rojiani of angiogenesis and lymphangiogenesis factors predicts
2002). In addition, preclinical investigations suggest outcome for non-small cell lung cancer patients. Virchows
that vascular disrupting approaches are likely to be Arch 458(3):331–340
complimentary to, rather than to duplicate anti- Anderson H, Yap JT et al (2003) Measurement of renal tumour
and normal tissue perfusion using positron emission
angiogenic strategies. Evidence suggests that anti- tomography in a phase II clinical trial of razoxane. Br J
angiogenic agents may be especially well suited for Cancer 89(2):262–267
micrometastatic disease or early-stage cancer (Lo- Angeletti CA, Lucchi M et al (1996) Prognostic significance of
zonschi et al. 1999), whereas vascular disrupting tumoral angiogenesis in completely resected late stage lung
carcinoma (stage IIIA-N2). Impact of adjuvant therapies in
agents may be particularly effective against large a subset of patients at high risk of recurrence. Cancer
bulky and late stage tumors (Landuyt et al. 2001; 78(3):409–415
Siemann and Shi 2003). Indeed data are beginning to Apolinario RM, van der Valk P et al (1997) Prognostic value of
emerge that combining these two strategies may the expression of p53, bcl-2, and bax oncoproteins, and
neovascularization in patients with radically resected non-
provide particularly beneficial therapeutic effects. small-cell lung cancer. J Clin Oncol 15(6):2456–2466
One current trial (NCT00653939) is designed to test Bach-Gansmo T, Bogsrud TV et al (2008) Integrin scintimam-
the efficacy of the combination of carboplatin, mography using a dedicated breast imaging, solid-state
34 W. Shi and D. W. Siemann

gamma-camera and (99m)Tc-labelled NC100692. Clin mRNA and endostatin mRNA in pleural effusions of patients
Physiol Funct Imaging 28(4):235–239 with lung cancer. Diagn Cytopathol (Epub ahead of print)
Baguley BC (2003) Antivascular therapy of cancer: DMXAA. Ching LM, Zwain S et al (2004) Relationship between tumour
Lancet Oncol 4(3):141–148 endothelial cell apoptosis and tumour blood flow shutdown
Baguley BC, McKeage MJ (2010) ASA404: a tumor vascular- following treatment with the antivascular agent DMXAA in
disrupting agent with broad potential for cancer therapy. mice. Br J Cancer 90(4):906–910
Future Oncol 6(10):1537–1543 Chouaib S, Kieda C et al (2010) Endothelial cells as key
Baguley BC, Siemann DW (2010) Temporal aspects of the determinants of the tumor microenvironment: interaction
action of ASA404 (vadimezan; DMXAA). Expert Opin with tumor cells, extracellular matrix and immune killer
Investig Drugs 19(11):1413–1425 cells. Crit Rev Immunol 30(6):529–545
Batchelor TT, Sorensen AG et al (2007) AZD2171, a pan- Ciardiello F, Caputo R et al (2003) Antitumor effects of
VEGF receptor tyrosine kinase inhibitor, normalizes tumor ZD6474, a small molecule vascular endothelial growth
vasculature and alleviates edema in glioblastoma patients. factor receptor tyrosine kinase inhibitor, with additional
Cancer Cell 11(1):83–95 activity against epidermal growth factor receptor tyrosine
Beadsmoore CJ, Screaton NJ (2003) Classification, staging and kinase. Clin Cancer Res 9(4):1546–1556
prognosis of lung cancer. Eur J Radiol 45(1):8–17 Clement-Duchene C, Wakelee H (2010) Antiangiogenic agents
Bergers G, Benjamin LE (2003) Tumorigenesis and the and vascular disrupting agents for the treatment of lung
angiogenic switch. Nat Rev Cancer 3(6):401–410 cancer: a review. J Thorac Oncol 5(1):129–139
Bernaards CHP, Chen D et al (2010) Circulating VEGF as a Collingridge DR, Carroll VA et al (2002) The development of
biomarker for bevacizumab based therapy in metastatic [(124)I]iodinated-VG76e: a novel tracer for imaging vas-
colorectal, non-small cell lung, and renal cell cancers: cular endothelial growth factor in vivo using positron
Analysis of phase III studies. J Clin Oncol 28(suppl):10519 emission tomography. Cancer Res 62(20):5912–5919
Bissett D, O’Byrne KJ et al (2005) Phase III study of matrix Comis RL (2003) A brief history of the research and treatment
metalloproteinase inhibitor prinomastat in non-small-cell of lung cancer from 1970 to 2003. Int J Clin Oncol
lung cancer. J Clin Oncol 23(4):842–849 8(4):230–233
Bonnesen B, Pappot H et al (2009) Vascular endothelial growth Connolly DT, Olander JV et al (1989) Human vascular
factor A and vascular endothelial growth factor receptor 2 permeability factor. Isolation from U937 cells. J Biol Chem
expression in non-small cell lung cancer patients: relation to 264(33):20017–20024
prognosis. Lung Cancer 66(3):314–318 D’Amato RJ, Loughnan MS et al (1994) Thalidomide is an
Bono AV, Celato N et al (2002) Microvessel density in prostate inhibitor of angiogenesis. Proc Natl Acad Sci U S A
carcinoma. Prostate Cancer Prostatic Dis 5(2):123–127 91(9):4082–4085
Botrel TE, Clark O et al (2011) Efficacy of bevacizumab (Bev) Dalaveris E, Kerenidi T et al (2009) VEGF, TNF-alpha and 8-
plus chemotherapy (CT) compared to CT alone in previ- isoprostane levels in exhaled breath condensate and serum
ously untreated locally advanced or metastatic non-small of patients with lung cancer. Lung Cancer 64(2):219–225
cell lung cancer (NSCLC): Systematic review and meta- Dannenberg AJ, Altorki NK et al (2001) Cyclo-oxygenase 2: a
analysis. Lung Cancer (Epub ahead of print) pharmacological target for the prevention of cancer. Lancet
Brattstrom D, Bergqvist M et al (1998) Basic fibroblast growth Oncol 2(9):544–551
factor and vascular endothelial growth factor in sera from de Boer RH, Arrieta O et al (2011) Vandetanib Plus Pemetr-
non-small-cell lung cancer patients. Anticancer Res exed for the Second-Line Treatment of Advanced Non-
18(2A):1123–1127 Small-Cell Lung Cancer: A Randomized, Double-Blind
Brooks PC, Montgomery AM et al (1994) Integrin alpha v beta Phase III Trial. J Clin Oncol (Epub ahead of print)
3 antagonists promote tumor regression by inducing apop- Decaussin M, Sartelet H et al (1999) Expression of vascular
tosis of angiogenic blood vessels. Cell 79(7):1157–1164 endothelial growth factor (VEGF) and its two receptors
Caron C, Spring K et al (2009) Non-redundant roles of the (VEGF-R1-Flt1 and VEGF-R2-Flk1/KDR) in non-small
Gab1 and Gab2 scaffolding adapters in VEGF-mediated cell lung carcinomas (NSCLCs): Correlation with angio-
signalling, migration, and survival of endothelial cells. Cell genesis and survival. J Pathol 188(4):369–377
Signal 21(6):943–953 Denekamp J (1990) Vascular attack as a therapeutic strategy for
Carrillo de Santa Pau E, Arias FC et al (2009) Prognostic cancer. Cancer Metastasis Rev 9(3):267–282
significance of the expression of vascular endothelial Deshpande N, Pysz MA et al (2010) Molecular ultrasound
growth factors A, B, C, and D and their receptors R1, R2, assessment of tumor angiogenesis. Angiogenesis 13(2):175–
and R3 in patients with non-small-cell lung cancer. Cancer 188
115(8):1701–1712 Dickinson AJ, Fox SB et al (1994) Quantification of angio-
Carrillo-de Santa Pau E, Carrillo Arias F et al (2010) Vascular genesis as an independent predictor of prognosis in invasive
endothelial growth factor (VEGF) serum levels are associ- bladder carcinomas. Br J Urol 74(6):762–766
ated with survival in early stages of lung cancer patients. Dowlati A, Subbiah S et al (2007) Phase II trial of thalidomide
Cancer Invest 28(4):393–398 as maintenance therapy for extensive stage small cell lung
Chandrachud LM, Pendleton N et al (1997) Relationship cancer after response to chemotherapy. Lung Cancer
between vascularity, age and survival in non-small-cell lung 56(3):377–381
cancer. Br J Cancer 76(10):1367–1375 Dowlati A, Gray R et al (2008) Cell adhesion molecules,
Chen Y, Liang B et al (2010) Transcription expression and vascular endothelial growth factor, and basic fibroblast
clinical significance of vascular endothelial growth factor growth factor in patients with non-small cell lung cancer
Angiogenesis and Lung Cancer 35

treated with chemotherapy with or without bevacizumab–an patients with advanced ovarian carcinoma. Anticancer Res
Eastern Cooperative Oncology Group Study. Clin Cancer 23(1B):549–556
Res 14(5):1407–1412 Galbraith SM, Chaplin DJ et al (2001) Effects of combretastatin
Drevs J (2003) Soluble markers for the detection of hypoxia A4 phosphate on endothelial cell morphology in vitro and
under antiangiogenic treatment. Anticancer Res relationship to tumour vascular targeting activity in vivo.
23(2A):1159–1161 Anticancer Res 21(1A):93–102
Dudek A, Gupta K et al (2010) Tumor angiogenesis. J Oncol Galbraith SM, Lodge MA et al (2002) Reproducibility of
(Epub ahead of print) dynamic contrast-enhanced MRI in human muscle and
Dudek AZ, Lesniewski-Kmak K et al (2009) Phase II trial of tumours: Comparison of quantitative and semi-quantitative
neoadjuvant therapy with carboplatin, gemcitabine plus analysis. NMR Biomed 15(2):132–142
thalidomide for stages IIB and III non-small-cell lung Galbraith SM, Maxwell RJ et al (2003) Combretastatin A4
cancer. J Thorac Oncol 4(8):969–975 phosphate has tumor antivascular activity in rat and man as
Edwards BK, Brown ML et al (2005) Annual report to the demonstrated by dynamic magnetic resonance imaging.
nation on the status of cancer, 1975–2002, featuring J Clin Oncol 21(15):2831–2842
population-based trends in cancer treatment. J Natl Cancer Galustian C, Dalgleish A (2009) Lenalidomide: a novel
Inst 97(19):1407–1427 anticancer drug with multiple modalities. Expert Opin
Eliceiri BP, Cheresh DA (1999) The role of alphav integrins Pharmacother 10(1):125–133
during angiogenesis: Insights into potential mechanisms of Gauler TC, BB Meric JB et al (2007) Phase II open-label study
action and clinical development. J Clin Invest 103(9):1227– to investigate efficacy and safety of PTK787/ZK222584
1230 orally administered once daily or twice daily at 1, 250 mg
Erdi YE, Macapinlac H et al (2000) Use of PET to monitor the as second line monotherapy in patients with stage IIIB/V
response of lung cancer to radiation treatment. Eur J Nucl NSCLC. J Clin Oncol 25:7541
Med 27(7):861–866 Gerber HP, Dixit V et al (1998) Vascular endothelial growth
Eriksson R, Persson HW et al (1991) Evaluation of Doppler factor induces expression of the antiapoptotic proteins Bcl-2
ultrasound for blood perfusion measurements. Ultrasound and A1 in vascular endothelial cells. J Biol Chem
Med Biol 17(5):445–452 273(21):13313–13316
Escudier B, Eisen T et al (2007) Sorafenib in advanced clear- Gialeli C, Theocharis AD et al (2011) Roles of matrix
cell renal-cell carcinoma. N Engl J Med 356(2):125–134 metalloproteinases in cancer progression and their pharma-
Fabbro D, Manley PW (2001) Su-6668. SUGEN. Curr Opin cological targeting. FEBS J 278(1):16–27
Investig Drugs 2(8):1142–1148 Gingras D, Boivin D et al (2003) Neovastat–a novel antiangio-
Feng Y, Wang W et al (2010) Expression of VEGF-C and genic drug for cancer therapy. Anticancer Drugs 14(2):91–96
VEGF-D as significant markers for assessment of lymphan- Goh V, Halligan S et al (2009) Can perfusion CT assessment of
giogenesis and lymph node metastasis in non-small-cell primary colorectal adenocarcinoma blood flow at staging
lung cancer. Anat Rec (Hoboken) 293(5):802–812 predict for subsequent metastatic disease? A pilot study. Eur
Ferrara N (2002) Role of vascular endothelial growth factor in Radiol 19(1):79–89
physiologic and pathologic angiogenesis: therapeutic impli- Gordon MS, Mendelson DS et al (2010) Tumor angiogenesis
cations. Semin Oncol 29(6 Suppl 16):10–14 and novel antiangiogenic strategies. Int J Cancer
Ferrara N, Gerber HP et al (2003) The biology of VEGF and its 126(8):1777–1787
receptors. Nat Med 9(6):669–676 Goss GD, Arnold A et al (2010) Randomized, double-blind trial
Folkman J (1971) Tumor angiogenesis: Therapeutic implica- of carboplatin and paclitaxel with either daily oral cediranib
tions. N Engl J Med 285(21):1182–1186 or placebo in advanced non-small-cell lung cancer: NCIC
Folkman J (1975) Tumor angiogenesis: a possible control point clinical trials group BR24 study. J Clin Oncol 28(1):49–55
in tumor growth. Ann Intern Med 82(1):96–100 Guddo F, Fontanini G et al (1999) The expression of basic
Folkman J (1990a) Endothelial cells and angiogenic growth fibroblast growth factor (bFGF) in tumor-associated stromal
factors in cancer growth and metastasis introduction. Cancer cells and vessels is inversely correlated with non-small-cell
Metastasis Rev 9(3):171–174 lung cancer progression. Hum Pathol 30(7):788–794
Folkman J (1990b) What is the evidence that tumors are Guidi AJ, Fischer L et al (1994) Microvessel density and
angiogenesis dependent? J Natl Cancer Inst 82(1):4–6 distribution in ductal carcinoma in situ of the breast. J Natl
Folkman J (1995) Angiogenesis in cancer, vascular, rheumatoid Cancer Inst 86(8):614–619
and other disease. Nat Med 1(1):27–31 Guo X, Chen Y et al (2009) Prognostic significance of VEGF-C
Folkman J (2002) Role of angiogenesis in tumor growth and expression in correlation with COX-2, lymphatic microves-
metastasis. Semin Oncol 29(6 Suppl 16):15–18 sel density, and clinicopathologic characteristics in human
Fontanini G, Bigini D et al (1995) Microvessel count predicts non-small-cell lung cancer. Acta Biochim Biophys Sin
metastatic disease and survival in non-small-cell lung (Shanghai) 41(3):217–222
cancer. J Pathol 177(1):57–63 Hasan J, Byers R et al (2002) Intra-tumoural microvessel density
Fontanini G, Faviana P et al (2002) A high vascular count and in human solid tumours. Br J Cancer 86(10):1566–1577
overexpression of vascular endothelial growth factor are Haubner R, Beer AJ et al (2010) Positron emission tomography
associated with unfavourable prognosis in operated small- tracers for imaging angiogenesis. Eur J Nucl Med Mol
cell-lung carcinoma. Br J Cancer 86(4):558–563 Imaging 37(Suppl 1):S86–103
Gadducci A, Viacava P et al (2003) Intratumoral microvessel Hawighorst H, Libicher M et al (1999) Evaluation of angio-
density, response to chemotherapy and clinical outcome of genesis and perfusion of bone marrow lesions: Role of
36 W. Shi and D. W. Siemann

semiquantitative and quantitative dynamic MRI. J Magn Jalal S, Bedano P et al (2010) Paclitaxel plus bevacizumab in
Reson Imaging 10(3):286–294 patients with chemosensitive relapsed small cell lung
Heng DY, Kollmannsberger C (2010) Sunitinib. Recent Results cancer: a safety, feasibility, and efficacy study from the
Cancer Res 184:71–82 Hoosier Oncology Group. J Thorac Oncol 5(12):2008–2011
Herbst RS, Hess KR et al (2002a) Phase I study of recombinant Jayson GC, Zweit J et al (2002) Molecular imaging and
human endostatin in patients with advanced solid tumors. biological evaluation of HuMV833 anti-VEGF antibody:
J Clin Oncol 20(18):3792–3803 Implications for trial design of antiangiogenic antibodies.
Herbst RS, Madden TL et al (2002b) Safety and pharmacoki- J Natl Cancer Inst 94(19):1484–1493
netic effects of TNP-470, an angiogenesis inhibitor, com- Jazieh AR, Komrokji R et al (2009) Phase II Trial of
bined with paclitaxel in patients with solid tumors: evidence thalidomide, irinotecan and gemcitabine in chemonaive
for activity in non-small-cell lung cancer. J Clin Oncol patients with advanced non-small cell lung cancer. Cancer
20(22):4440–4447 Invest: 27(9):923–926
Herbst RS, Sun Y et al (2010) Vandetanib plus docetaxel versus Jemal A, Siegel R et al (2010) Cancer statistics (2010). CA
docetaxel as second-line treatment for patients with Cancer J Clin 60(5):277–300
advanced non-small-cell lung cancer (ZODIAC): a dou- Johnson DH, Fehrenbacher L et al (2004) Randomized phase II
ble-blind, randomised, phase 3 trial. Lancet Oncol trial comparing bevacizumab plus carboplatin and paclitaxel
11(7):619–626 with carboplatin and paclitaxel alone in previously
Herrmann E, Bogemann M et al (2007) The role of the untreated locally advanced or metastatic non-small-cell
endothelin axis and microvessel density in bladder cancer— lung cancer. J Clin Oncol 22(11):2184–2191
correlation with tumor angiogenesis and clinical prognosis. Jubb AM, Harris AL (2010) Biomarkers to predict the clinical
Oncol Rep 18(1):133–138 efficacy of bevacizumab in cancer. Lancet Oncol
Heymach JV, Paz-Ares L et al (2008) Randomized phase II 11(12):1172–1183
study of vandetanib alone or with paclitaxel and carboplatin Kalmadi S, Davis M et al (2007) Phase I trial of three-weekly
as first-line treatment for advanced non-small-cell lung docetaxel, carboplatin and oral lenalidomide (Revlimid) in
cancer. J Clin Oncol 26(33):5407–5415 patients with advanced solid tumors. Invest New Drugs
Hidalgo M, Eckhardt SG (2001) Development of matrix 25(3):211–216
metalloproteinase inhibitors in cancer therapy. J Natl Can- Kanthou C, Tozer GM (2002) The tumor vascular targeting
cer Inst 93(3):178–193 agent combretastatin A-4-phosphate induces reorganization
Hlatky L, Hahnfeldt P et al (2002) Clinical application of of the actin cytoskeleton and early membrane blebbing in
antiangiogenic therapy: microvessel density, what it does human endothelial cells. Blood 99(6):2060–2069
and doesn’t tell us. J Natl Cancer Inst 94(12):883–893 Keith RL, Miller YE et al (2000) Angiogenic squamous
Hoekstra CJ, Stroobants SG et al (2002) Measurement of dysplasia in bronchi of individuals at high risk for lung
perfusion in stage IIIA-N2 non-small cell lung cancer using cancer. Clin Cancer Res 6(5):1616–1625
H(2)(15)O and positron emission tomography. Clin Cancer Kelly RJ, Rixe O (2010) Axitinib (AG-013736). Recent Results
Res 8(7):2109–2115 Cancer Res 184:33–44
Holash J, Maisonpierre PC et al (1999) Vessel cooption, Kepka L, Sprawka A et al (2009) Combination of radiotherapy
regression, and growth in tumors mediated by angiopoietins and chemotherapy in locally advanced NSCLC. Expert Rev
and VEGF. Science 284(5422):1994–1998 Anticancer Ther 9(10):1389–1403
Holden SN, Eckhardt SG et al (2005) Clinical evaluation of Kerbel RS (2000) Tumor angiogenesis: Past, present and the
ZD6474, an orally active inhibitor of VEGF and EGF near future. Carcinogenesis 21(3):505–515
receptor signaling, in patients with solid, malignant tumors. Kerbel RS (2008) Tumor angiogenesis. N Engl J Med
Ann Oncol 16(8):1391–1397 358(19):2039–2049
Horn L, Dahlberg SE et al (2009) Phase II study of cisplatin Khan AW, Dhillon AP et al (2002) Prognostic significance of
plus etoposide and bevacizumab for previously untreated, intratumoural microvessel density (IMD) in resected
extensive-stage small-cell lung cancer: Eastern Cooperative pancreatic and ampullary cancers to standard histopathol-
Oncology Group Study E3501. J Clin Oncol 27(35):6006– ogical variables and survival. Eur J Surg Oncol 28(6):637–
6011 644
Hu-Lowe DD, Zou HY et al (2008) Nonclinical antiangiogen- Kim KJ, Li B et al (1993) Inhibition of vascular endothelial
esis and antitumor activities of axitinib (AG-013736), an growth factor-induced angiogenesis suppresses tumour
oral, potent, and selective inhibitor of vascular endothelial growth in vivo. Nature 362(6423):841–844
growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Kostakoglu L, Goldsmith SJ (2003) 18F-FDG PET evaluation
Res 14(22):7272–7283 of the response to therapy for lymphoma and for breast,
Hurwitz H, Fehrenbacher L et al (2004) Bevacizumab plus lung, and colorectal carcinoma. J Nucl Med 44(2):224–
irinotecan, fluorouracil, and leucovorin for metastatic colo- 239
rectal cancer. N Engl J Med 350(23):2335–2342 Koukourakis MI, Giatromanolaki A et al (2000) Vascular
Hurwitz H, Douglas PS, Middleton JP et al (2010) Analysis of endothelial growth factor/KDR activated microvessel den-
early hypertension and clinical outcome with bevacizumab. sity versus CD31 standard microvessel density in non-
J Clin Oncol 28(suppl):3039 small-cell lung cancer. Cancer Res 60(11):3088–3095
Jain RK, Duda DG et al (2009) Biomarkers of response and Lacy MQ, Tefferi A (2011) Pomalidomide therapy for multiple
resistance to antiangiogenic therapy. Nat Rev Clin Oncol myeloma and myelofibrosis: an update. Leuk Lymphoma
6(6):327–338 52(4):560–566
Angiogenesis and Lung Cancer 37

Landuyt W, Theys J et al (2001) Effect of TNP-470 (AGM- Lopez-Campos JL, Sanchez Silva R et al (2011) Overexpres-
1470) on the growth of rat rhabdomyosarcoma tumors of sion of Aquaporin-1 in lung adenocarcinomas and pleural
different sizes. Cancer Invest 19(1):35–40 mesotheliomas. Histol Histopathol 26(4):451–459
Laurie SA, Gauthier I et al (2008) Phase I and pharmacokinetic Loupakis F, Falcone A et al (2007) Vascular endothelial growth
study of daily oral AZD2171, an inhibitor of vascular factor levels in immunodepleted plasma of cancer patients
endothelial growth factor tyrosine kinases, in combination as a possible pharmacodynamic marker for bevacizumab
with carboplatin and paclitaxel in patients with advanced activity. J Clin Oncol 25(13):1816–1818
non-small-cell lung cancer: the National Cancer Institute of Lozonschi L, Sunamura M et al (1999) Controlling tumor
Canada clinical trials group. J Clin Oncol 26(11):1871– angiogenesis and metastasis of C26 murine colon adeno-
1878 carcinoma by a new matrix metalloproteinase inhibitor,
Lee JS, Kim HS et al (2002) Expression of vascular endothelial KB-R7785, in two tumor models. Cancer Res 59(6):1252–
growth factor in adenocarcinomas of the uterine cervix and 1258
its relation to angiogenesis and p53 and c-erbB-2 protein Lu JF, Bruno R et al (2008) Clinical pharmacokinetics of
expression. Gynecol Oncol 85(3):469–475 bevacizumab in patients with solid tumors. Cancer Chemo-
Lee SM, Rudd R et al (2009) Randomized double-blind ther Pharmacol 62(5):779–786
placebo-controlled trial of thalidomide in combination with Lu C, Lee JJ et al (2010) Chemoradiotherapy with or without
gemcitabine and Carboplatin in advanced non-small-cell AE-941 in stage III non-small-cell lung cancer: a random-
lung cancer. J Clin Oncol 27(31):5248–5254 ized phase III trial. J Natl Cancer Inst 102(12):859–865
Leighl NB, Paz-Ares L et al (2005) Randomized phase III study Lucchi M, Mussi A et al (2002) Small-cell lung carcinoma
of matrix metalloproteinase inhibitor BMS–275291 in (SCLC): the angiogenic phenomenon. Eur J Cardiothorac
combination with paclitaxel and carboplatin in advanced Surg 21(6):1105–1110
non-small-cell lung cancer: National Cancer Institute of Macchiarini P, Fontanini G et al (1992) Relation of neovascu-
Canada-Clinical Trials Group Study BR. 18. J Clin Oncol larisation to metastasis of non-small-cell lung cancer.
23(12):2831–2839 Lancet 340(8812):145–146
Leighl NB, Raez LE et al (2010) A multicenter, phase 2 study Macluskey M, Baillie R et al (2000) High levels of apoptosis
of vascular endothelial growth factor trap (Aflibercept) in are associated with improved survival in non-small cell lung
platinum- and erlotinib-resistant adenocarcinoma of the cancer. Anticancer Res 20(3B):2123–2128
lung. J Thorac Oncol 5(7):1054–1059 Mahzouni P, Mohammadizadeh F et al (2010) Determining the
Leong-Poi H, Christiansen J et al (2003) Noninvasive relationship between ‘‘microvessel density and different
assessment of angiogenesis by ultrasound and microbub- grades of astrocytoma based on immunohistochemistry for
bles targeted to alpha(v)-integrins. Circulation 107(3):455– factor VIII-related antigen (von Willebrand factor) expres-
460 sion in tumor microvessels. Indian J Pathol Microbiol
Levitt NC, Eskens FA et al (2001) Phase I and pharmacological 53(4):605–610
study of the oral matrix metalloproteinase inhibitor, Massi D, Franchi A et al (2002) Tumor angiogenesis as a
MMI270 (CGS27023A), in patients with advanced solid prognostic factor in thick cutaneous malignant melanoma. A
cancer. Clin Cancer Res 7(7):1912–1922 quantitative morphologic analysis. Virchows Arch
Li X, Liu X et al (2003) Thalidomide down-regulates the 440(1):22–28
expression of VEGF and bFGF in cisplatin-resistant human Matsuyama W, Hashiguchi T et al (2000) Serum levels of
lung carcinoma cells. Anticancer Res 23(3B):2481–2487 vascular endothelial growth factor dependent on the stage
Li KL, Wilmes LJ et al (2005) Heterogeneity in the progression of lung cancer. Chest 118(4):948–951
angiogenic response of a BT474 human breast cancer to Mattern J, Koomagi R et al (1996) Association of vascular
a novel vascular endothelial growth factor-receptor tyro- endothelial growth factor expression with intratumoral
sine kinase inhibitor: assessment by voxel analysis of microvessel density and tumour cell proliferation in human
dynamic contrast-enhanced MRI. J Magn Reson Imaging epidermoid lung carcinoma. Br J Cancer 73(7):931–934
22(4):511–519 McCulloch P, Choy A et al (1995) Association between tumour
Liekens S, De Clercq E et al (2001) Angiogenesis: Regulators angiogenesis and tumour cell shedding into effluent venous
and clinical applications. Biochem Pharmacol 61(3):253– blood during breast cancer surgery. Lancet
270 346(8986):1334–1335
Lin Q, Li M et al (2010) Prognostic impact of vascular McKeage MJ, Reck M et al (2009) Phase II study of ASA404
endothelial growth factor-A and E-cadherin expression in (vadimezan, 5, 6-dimethylxanthenone-4-acetic acid/
completely resected pathologic stage I non-small-cell lung DMXAA) 1800mg/m(2) combined with carboplatin and
cancer. Jpn J Clin Oncol 40(7):670–676 paclitaxel in previously untreated advanced non-small-cell
Ling Y, Yang Y et al (2007) Endostar, a novel recombinant lung cancer. Lung Cancer 65(2):192–197
human endostatin, exerts antiangiogenic effect via blocking Medetoglu B, Gunluoglu MZ et al (2010) Tumor angiogenesis
VEGF-induced tyrosine phosphorylation of KDR/Flk-1 of in predicting the survival of patients with stage I lung
endothelial cells. Biochem Biophys Res Commun cancer. J Thorac Cardiovasc Surg 140(5):996–1000
361(1):79–84 Miettinen M (1993) Immunohistochemistry in tumour diagno-
Lockhart AC, Rothenberg ML et al (2010) Phase I study of sis. Ann Med 25(3):221–233
intravenous vascular endothelial growth factor trap, afliber- Mihaylova Z, Ludovini V et al (2007) Serum level changes of
cept, in patients with advanced solid tumors. J Clin Oncol matrix metalloproteinases 2 and 9, vascular endothelial
28(2):207–214 growth factor and epidermal growth factor receptor during
38 W. Shi and D. W. Siemann

platinum-based chemotherapy in advanced non-small-cell Nilsson F, Kosmehl H et al (2001) Targeted delivery of tissue
lung cancer patients. J BUON 12(1):105–111 factor to the ED-B domain of fibronectin, a marker of
Miles KA (2003) Perfusion CT for the assessment of tumour angiogenesis, mediates the infarction of solid tumors in
vascularity: Which protocol? Br J Radiol 76(Spec No mice. Cancer Res 61(2):711–716
1):S36–42 Novello S, Scagliotti GV et al (2009) Phase II study of
Miles KA, Charnsangavej C et al (2000) Application of CT in continuous daily sunitinib dosing in patients with previously
the investigation of angiogenesis in oncology. Acad Radiol treated advanced non-small-cell lung cancer. Br J Cancer
7(10):840–850 101(9):1543–1548
Miller VA, O’CP, Soh C et al (2009) A randomized double- O’Reilly MS (1997) Angiostatin: an endogenous inhibitor of
blind, placebo controlled, phase IIIb trial (ATLAS) angiogenesis and of tumor growth. EXS 79:273–294
comparing bevacizumab (B) therapy with or without O’Reilly MS, Holmgren L et al (1994) Angiostatin: a circu-
erlotinib (E) after completeion of chemotherapy with B lating endothelial cell inhibitor that suppresses angiogenesis
for first-line tratment of locally advanced, recurrent, or and tumor growth. Cold Spring Harb Symp Quant Biol
metastastic non-small-cell lung cancer (NSCLC). J Clin 59:471–482
Oncol 27(18s) O’Reilly MS, Boehm T et al (1997) Endostatin: an endogenous
Miller AA, Case D et al (2006) Phase II study of carboplatin, inhibitor of angiogenesis and tumor growth. Cell 88(2):277–
irinotecan, and thalidomide in patients with advanced non- 285
small-cell lung cancer. J Thorac Oncol 1(8):832–836 Ohta Y, Endo Y et al (1996) Significance of vascular
Miller AA, Case D et al (2007) Phase I study of lenalidomide in endothelial growth factor messenger RNA expression in
solid tumors. J Thorac Oncol 2(5):445–449 primary lung cancer. Clin Cancer Res 2(8):1411–1416
Minardi D, Lucarini G et al (2008) Prognostic role of tumor Ohta Y, Ohta N et al (2002) Vascular endothelial growth factor
necrosis, microvessel density, vascular endothelial growth expression in airways of patients with lung cancer: a
factor and hypoxia inducible factor-1alpha in patients with possible diagnostic tool of responsive angiogenic status on
clear cell renal carcinoma after radical nephrectomy in a the host side. Chest 121(5):1624–1627
long term follow-up. Int J Immunopathol Pharmacol Oshika Y, Nakamura M et al (2000) Ribozyme approach to
21(2):447–455 downregulate vascular endothelial growth factor (VEGF)
Moore BB, Arenberg DA et al (1998) Tumor angiogenesis is 189 expression in non-small-cell lung cancer (NSCLC). Eur
regulated by CXC chemokines. J Lab Clin Med 132(2):97– J Cancer 36(18):2390–2396
103 Osterlund P, Soveri LM et al (2011) Hypertension and overall
Mountain CF (2000) The international system for staging lung survival in metastatic colorectal cancer patients treated with
cancer. Semin Surg Oncol 18(2):106–115 bevacizumab-containing chemotherapy. Br J Cancer
Mountain CF, Hermes KE (2003) Surgical treatment of lung 104(4):599–604
cancer. Past and present. Methods Mol Med 75:453–487 Paku S (1998) Current concepts of tumor-induced angiogenesis.
Mross K, Stefanic M et al (2010) Phase I study of the Pathol Oncol Res 4(1):62–75
angiogenesis inhibitor BIBF 1120 in patients with advanced Papamichael D (2001) Prognostic role of angiogenesis in
solid tumors. Clin Cancer Res 16(1):311–319 colorectal cancer. Anticancer Res 21(6B):4349–4353
Mutter R, Lu B et al (2009) A phase II study of celecoxib in Papapetropoulos A, Garcia-Cardena G et al (1999) Direct
combination with paclitaxel, carboplatin, and radiotherapy actions of angiopoietin-1 on human endothelium: Evidence
for patients with inoperable stage IIIA/B non-small-cell for network stabilization, cell survival, and interaction with
lung cancer. Clin Cancer Res 15(6):2158–2165 other angiogenic growth factors. Lab Invest 79(2):213–223
Natale RB, Thongprasert S et al (2011) Phase III trial of Park MS, Klotz E et al (2009) Perfusion CT: Noninvasive
vandetanib compared with erlotinib in patients with previ- surrogate marker for stratification of pancreatic cancer
ously treated advanced non-small-cell lung cancer. J Clin response to concurrent chemo- and radiation therapy.
Oncol (Epub ahead of print) Radiology 250(1):110–117
Natale RB, Bodkin D et al (2009) Vandetanib versus gefitinib Pastorino U, Andreola S et al (1997) Immunocytochemical
in patients with advanced non-small-cell lung cancer: markers in stage I lung cancer: Relevance to prognosis.
results from a two-part, double-blind, randomized phase ii J Clin Oncol 15(8):2858–2865
study. J Clin Oncol 27(15):2523–2529 Patel JD, Hensing TA et al (2009) Phase II study of pemetrexed
Ng QS, Goh V et al (2007a) Tumor antivascular effects of and carboplatin plus bevacizumab with maintenance
radiotherapy combined with combretastatin a4 phosphate in pemetrexed and bevacizumab as first-line therapy for
human non-small-cell lung cancer. Int J Radiat Oncol Biol nonsquamous non-small-cell-lung cancer. J Clin Oncol
Phys 67(5):1375–1380 27(20):3284–3289
Ng QS, Goh V et al (2007b) Acute tumor vascular effects Pathak AP, Penet MF et al (2010) MR molecular imaging of
following fractionated radiotherapy in human lung cancer: tumor vasculature and vascular targets. Adv Genet 69:1–30
In vivo whole tumor assessment using volumetric perfusion Pavco PA, Bouhana KS et al (2000) Antitumor and antimet-
computed tomography. Int J Radiat Oncol Biol Phys astatic activity of ribozymes targeting the messenger RNA
67(2):417–424 of vascular endothelial growth factor receptors. Clin Cancer
Ng CS, Kodama Y et al (2009) Tumor blood flow measured by Res 6(5):2094–2103
perfusion computed tomography and 15O-labeled water Pezzella F, Pastorino U et al (1997) Non-small-cell lung
positron emission tomography: a comparison study. J Com- carcinoma tumor growth without morphological evidence of
put Assist Tomogr 33(3):460–465 neo-angiogenesis. Am J Pathol 151(5):1417–1423
Angiogenesis and Lung Cancer 39

Philpott M, Joseph WR et al (1997) Production of tumour advanced solid tumors: Pharmacokinetic and clinical
necrosis factor-alpha by cultured human peripheral blood results. J Clin Oncol 23(24):5474–5483
leucocytes in response to the anti-tumour agent 5, 6- Ruotsalainen T, Joensuu H et al (2002) High pretreatment
dimethylxanthenone-4-acetic acid (NSC 640488). Br J serum concentration of basic fibroblast growth factor is a
Cancer 76(12):1586–1591 predictor of poor prognosis in small-cell-lung cancer.
Polverino A, Coxon A et al (2006) AMG 706, an oral, Cancer Epidemiol Biomarkers Prev 11(11):1492–1495
multikinase inhibitor that selectively targets vascular endo- Rustin GJ, Shreeves G et al (2010) A Phase Ib trial of CA4P
thelial growth factor, platelet-derived growth factor, and kit (combretastatin A-4 phosphate), carboplatin, and paclitaxel
receptors, potently inhibits angiogenesis and induces regres- in patients with advanced cancer. Br J Cancer 102(9):1355–
sion in tumor xenografts. Cancer Res 66(17):8715–8721 1360
Posther KE, Harpole DH Jr (2006) The surgical management of Salven P, Ruotsalainen T et al (1998) High pre-treatment serum
lung cancer. Cancer Invest 24(1):56–67 level of vascular endothelial growth factor (VEGF) is
Poulaki V (2002) BMS-275291. Bristol-Myers Squibb. Curr associated with poor outcome in small-cell lung cancer. Int
Opin Investig Drugs 3(3):500–504 J Cancer 79(2):144–146
Purdie TG, Henderson E et al (2001) Functional CT imaging of Sandler A, Gray R et al (2006) Paclitaxel-carboplatin alone or
angiogenesis in rabbit VX2 soft-tissue tumour. Phys Med with bevacizumab for non-small-cell lung cancer. N Engl J
Biol 46(12):3161–3175 Med 355(24):2542–2550
Rades D, Setter C et al (2010) Fibroblast growth factor 2-A Scagliotti G, Novello S et al (2010) Phase III study of
predictor of outcome for patients irradiated for stage ii-iii carboplatin and paclitaxel alone or with sorafenib in
non-small-cell lung cancer. Int J Radiat Oncol Biol Phys advanced non-small-cell lung cancer. J Clin Oncol
(Epub ahead of print) 28(11):1835–1842
Rades D, Setter C et al (2010) Prognostic impact of VEGF and Schiller JH, Larson T et al (2009) Efficacy and safety of axitinib
VEGF receptor 1 (FLT1) expression in patients irradiated in patients with advanced non-small-cell lung cancer:
for stage II/III non-small cell lung cancer (NSCLC). Results from a phase II study. J Clin Oncol 27(23):3836–
Strahlenther Onkol 186(6):307–314 3841
Reck M, Kaiser R et al (2011) A phase II double-blind study to Schneider BJ, Kalemkerian GP et al (2008a) Phase II study of
investigate efficacy and safety of two doses of the triple celecoxib and docetaxel in non-small cell lung cancer
angiokinase inhibitor BIBF 1120 in patients with relapsed (NSCLC) patients with progression after platinum-based
advanced non-small-cell lung cancer. Ann Oncol therapy. J Thorac Oncol 3(12):1454–1459
Reck M, von Pawel J et al (2009) Phase III trial of cisplatin plus Schneider BP, Wang M et al (2008b) Association of vascular
gemcitabine with either placebo or bevacizumab as first-line endothelial growth factor, vascular endothelial growth
therapy for nonsquamous non-small-cell lung cancer: factor receptor-2 genetic polymorphisms with outcome in
AVAil. J Clin Oncol 27(8):1227–1234 a trial of paclitaxel compared with paclitaxel plus bev-
Reckamp K, Gitlitz B et al (2011) Biomarker-based phase I acizumab in advanced breast cancer: ECOG 2100. J Clin
dose-escalation, pharmacokinetic, and pharmacodynamic Oncol 26(28):4672–4678
study of oral apricoxib in combination with erlotinib in Schnell O, Krebs B et al (2009) Imaging of integrin
advanced nonsmall cell lung cancer. Cancer 117(4):809– alpha(v)beta(3) expression in patients with malignant
818 glioma by [18F] Galacto-RGD positron emission tomogra-
Riely GJ, Miller VA (2007) Vascular endothelial growth factor phy. Neuro Oncol 11(6):861–870
trap in non-small-cell lung cancer. Clin Cancer Res 13(15 Pt Schultheis AM, Lurje G et al (2008) Polymorphisms and
2):s4623–4627 clinical outcome in recurrent ovarian cancer treated with
Rimassa L, Santoro A (2009) Sorafenib therapy in advanced cyclophosphamide and bevacizumab. Clin Cancer Res
hepatocellular carcinoma: the SHARP trial. Expert Rev 14(22):7554–7563
Anticancer Ther 9(6):739–745 Schwartz JD, Rowinsky EK et al (2010) Vascular endothelial
Rini BI, Halabi S et al (2010) Phase III trial of bevacizumab growth factor receptor-1 in human cancer: concise review
plus interferon alfa versus interferon alfa monotherapy in and rationale for development of IMC-18F1 (Human
patients with metastatic renal cell carcinoma: final results of antibody targeting vascular endothelial growth factor
CALGB 90206. J Clin Oncol 28(13):2137–2143 receptor-1). Cancer 116(4 Suppl):1027–1032
Risau W (1997) Mechanisms of angiogenesis. Nature Scott EN, Meinhardt G et al (2007) Vatalanib: the clinical
386(6626):671–674 development of a tyrosine kinase inhibitor of angiogenesis
Roca C, Adams RH (2007) Regulation of vascular morpho- in solid tumours. Expert Opin Investig Drugs 16(3):367–379
genesis by Notch signaling. Genes Dev 21(20):2511–2524 Senger DR, Perruzzi CA et al (1986) A highly conserved
Roodink I, Leenders WP (2010) Targeted therapies of cancer: vascular permeability factor secreted by a variety of human
angiogenesis inhibition seems not enough. Cancer Lett and rodent tumor cell lines. Cancer Res 46(11):5629–5632
299(1):1–10 Seto T, Higashiyama M et al (2006) Prognostic value of
Rosen LS, Kurzrock R et al (2007) Safety, pharmacokinetics, expression of vascular endothelial growth factor and its flt-1
and efficacy of AMG 706, an oral multikinase inhibitor, in and KDR receptors in stage I non-small-cell lung cancer.
patients with advanced solid tumors. J Clin Oncol Lung Cancer 53(1):91–96
25(17):2369–2376 Shepherd FA, Giaccone G et al (2002) Prospective, random-
Rugo HS, Herbst RS et al (2005) Phase I trial of the oral ized, double-blind, placebo-controlled trial of marimastat
antiangiogenesis agent AG-013736 in patients with after response to first-line chemotherapy in patients with
40 W. Shi and D. W. Siemann

small-cell lung cancer: a trial of the National Cancer Swensen SJ, Yamashita K et al (2000) Lung nodules: Dual-
Institute of Canada-Clinical Trials Group and the European kilovolt peak analysis with CT–multicenter study. Radiol-
Organization for Research and Treatment of Cancer. J Clin ogy 214(1):81–85
Oncol 20(22):4434–4439 Takahashi S, Shinya T et al (2010) Angiostatin inhibition of
Shi W, Siemann DW (2002) Inhibition of renal cell carcinoma vascular endothelial growth factor-stimulated nitric oxide
angiogenesis and growth by antisense oligonucleotides production in endothelial cells. J Pharmacol Sci
targeting vascular endothelial growth factor. Br J Cancer 112(4):432–437
87(1):119–126 Takase Y, Kai K et al (2010) Endoglin (CD105) expression and
Siemann DW (2011) The unique characteristics of tumor angiogenesis status in small-cell lung cancer. Pathol Res
vasculature and preclinical evidence for its selective Pract 206(11):725–730
disruption by tumor-vascular disrupting agents. Cancer Takigawa N, Segawa Y et al (1998) Elevated vascular
Treat Rev 37(1):63–74 endothelial growth factor levels in sera of patients with
Siemann DW, Horsman MR (2002) Enhancement of radiation lung cancer. Anticancer Res 18(2B):1251–1254
therapy by vascular targeting agents. Curr Opin Investig Tamura M, Ohta Y et al (2001) Plasma VEGF concentration
Drugs 3(11):1660–1665 can predict the tumor angiogenic capacity in non-small-cell
Siemann DW, Rojiani AM (2002) Antitumor efficacy of lung cancer. Oncol Rep 8(5):1097–1102
conventional anticancer drugs is enhanced by the vascular Tamura M, Ohta Y et al (2002) Diagnostic value of plasma
targeting agent ZD6126. Int J Radiat Oncol Biol Phys vascular endothelial growth factor as a tumor marker in
54(5):1512–1517 patients with non-small-cell lung cancer. Int J Biol Markers
Siemann DW, Shi W (2003) Targeting the tumor blood vessel 17(4):275–279
network to enhance the efficacy of radiation therapy. Semin Tanigawa N, Amaya H et al (1996) Extent of tumor vascular-
Radiat Oncol 13(1):53–61 ization correlates with prognosis and hematogenous metas-
Sin N, Meng L et al (1997) The anti-angiogenic agent tasis in gastric carcinomas. Cancer Res 56(11):2671–2676
fumagillin covalently binds and inhibits the methionine Tateishi U, Kusumoto M et al (2002) Contrast-enhanced
aminopeptidase, MetAP-2. Proc Natl Acad Sci U S A dynamic computed tomography for the evaluation of tumor
94(12):6099–6103 angiogenesis in patients with lung carcinoma. Cancer
Socinski MA, Novello S et al (2008) Multicenter, phase II trial 95(4):835–842
of sunitinib in previously treated, advanced non-small-cell Teng LS, Jin KT et al (2010) Clinical applications of VEGF-
lung cancer. J Clin Oncol 26(4):650–656 trap (aflibercept) in cancer treatment. J Chin Med Assoc
Socinski MA, Langer CJ et al (2009) Safety of bevacizumab in 73(9):449–456
patients with non-small-cell lung cancer and brain metas- Thomas JP, Arzoomanian RZ et al (2003) Phase I pharmaco-
tases. J Clin Oncol 27(31):5255–5261 kinetic and pharmacodynamic study of recombinant human
Solorzano CC, Baker CH et al (2001) Inhibition of growth and endostatin in patients with advanced solid tumors. J Clin
metastasis of human pancreatic cancer growing in nude Oncol 21(2):223–231
mice by PTK 787/ZK222584, an inhibitor of the vascular Toi M, Kashitani J et al (1993) Tumor angiogenesis is an
endothelial growth factor receptor tyrosine kinases. Cancer independent prognostic indicator in primary breast carci-
Biother Radiopharm 16(5):359–370 noma. Int J Cancer 55(3):371–374
Spigel DR, Greco FA et al (2009) Phase II trial of irinotecan, Trepel M, Grifman M et al (2000) Molecular adaptors for
carboplatin, and bevacizumab in the treatment of patients vascular-targeted adenoviral gene delivery. Hum Gene Ther
with extensive-stage small-cell lung cancer. J Thorac Oncol 11(14):1971–1981
4(12):1555–1560 Trivella M, Pezzella F et al (2007) Microvessel density as a
Spratlin JL, Cohen RB et al (2010) Phase I pharmacologic and prognostic factor in non-small-cell lung carcinoma: a meta-
biologic study of ramucirumab (IMC-1121B), a fully human analysis of individual patient data. Lancet Oncol 8(6):488–
immunoglobulin G1 monoclonal antibody targeting the 499
vascular endothelial growth factor receptor-2. J Clin Oncol Tuder RM, Cool CD et al (2001) The pathobiology of
28(5):780–787 pulmonary hypertension. Endothelium. Clin Chest Med
St Croix B, Rago C et al (2000) Genes expressed in human 22(3):405–418
tumor endothelium. Science 289(5482):1197–1202 Ucuzian AA, Gassman AA et al (2010) Molecular mediators of
Sternberg CN, Davis ID et al (2010) Pazopanib in locally angiogenesis. J Burn Care Res 31(1):158–175
advanced or metastatic renal cell carcinoma: Results of a Ueno K, Inoue Y et al (2001) Increased serum levels of basic
randomized phase III trial. J Clin Oncol 28(6):1061–1068 fibroblast growth factor in lung cancer patients: Relevance
Steward WP (1999) Marimastat (BB2516): Current status of to response of therapy and prognosis. Lung Cancer 31(2–
development. Cancer Chemother Pharmacol 43(Sup- 3):213–219
pl):S56–60 van der Woude HJ, Bloem JL et al (1995) Treatment of high-
Strizzi L, Vianale G et al (2001) Basic fibroblast growth grade bone sarcomas with neoadjuvant chemotherapy: the
factor in mesothelioma pleural effusions: Correlation with utility of sequential color Doppler sonography in predicting
patient survival and angiogenesis. Int J Oncol 18(5):1093– histopathologic response. Am J Roentgenol 165(1):125–133
1098 Varallyay CG, Muldoon LL et al (2009) Dynamic MRI using
Strobel D, Krodel U et al (2000) Clinical evaluation of contrast- iron oxide nanoparticles to assess early vascular effects of
enhanced color Doppler sonography in the differential antiangiogenic versus corticosteroid treatment in a glioma
diagnosis of liver tumors. J Clin Ultrasound 28(1):1–13 model. J Cereb Blood Flow Metab 29(4):853–860
Angiogenesis and Lung Cancer 41

Veenendaal LM, Jin H et al (2002) In vitro and in vivo studies WHO (1979) WHO handbook for reporting results of cancer
of a VEGF121/rGelonin chimeric fusion toxin targeting the treatment. World Health Organization. Geneva, Switzerland
neovasculature of solid tumors. Proc Natl Acad Sci U S A Willett CG, Duda DG et al (2009) Efficacy, safety, and
99(12):7866–7871 biomarkers of neoadjuvant bevacizumab, radiation therapy,
Vermeulen PB, Gasparini G et al (1996) Quantification of and fluorouracil in rectal cancer: a multidisciplinary phase II
angiogenesis in solid human tumours: an international study. J Clin Oncol 27(18):3020–3026
consensus on the methodology and criteria of evaluation. Witte L, Hicklin DJ et al (1998) Monoclonal antibodies
Eur J Cancer 32A(14):2474–2484 targeting the VEGF receptor-2 (Flk1/KDR) as an anti-
Vihinen P, Kahari VM (2002) Matrix metalloproteinases in angiogenic therapeutic strategy. Cancer Metastasis Rev
cancer: prognostic markers and therapeutic targets. Int J 17(2):155–161
Cancer 99(2):157–166 Wu XH, Qian C et al (2011) Correlations of hypoxia-inducible
Volm M, Koomagi R et al (1997) Prognostic value of basic factor-1alpha/hypoxia-inducible factor-2alpha expression
fibroblast growth factor and its receptor (FGFR-1) in with angiogenesis factors expression and prognosis in
patients with non-small-cell lung carcinomas. Eur J Cancer non-small-cell lung cancer. Chin Med J (Engl) 124(1):11–
33(4):691–693 18
Watanabe Y, Dvorak HF (1997) Vascular permeability factor/ Yamashita J, Ogawa M et al (1999) Platelet-derived endothelial
vascular endothelial growth factor inhibits anchorage- cell growth factor/thymidine phosphorylase concentrations
disruption-induced apoptosis in microvessel endothelial differ in small cell and non-small-cell lung cancer. Chest
cells by inducing scaffold formation. Exp Cell Res 116(1):206–211
233(2):340–349 Yamazaki K, Abe S et al (1994) Tumor angiogenesis in human
Webb NJ, Bottomley MJ et al (1998) Vascular endothelial lung adenocarcinoma. Cancer 74(8):2245–2250
growth factor (VEGF) is released from platelets during Yang DX, Li NE et al (2010) Expression of Elf-1 and survivin
blood clotting: implications for measurement of circulating in non-small cell lung cancer and their relationship to
VEGF levels in clinical disease. Clin Sci (Lond) 94(4):395– intratumoral microvessel density. Chin J Cancer 29(4):396–
404 402
Wedam SB, Low JA et al (2006) Antiangiogenic and antitumor Yi CA, Lee KS et al (2004) Solitary pulmonary nodules:
effects of bevacizumab in patients with inflammatory and dynamic enhanced multi-detector row CT study and com-
locally advanced breast cancer. J Clin Oncol 24(5):769–777 parison with vascular endothelial growth factor and micro-
Wedge SR, Kendrew J et al (2005) AZD2171: a highly potent, vessel density. Radiology 233(1):191–199
orally bioavailable, vascular endothelial growth factor Zahiragic L, Schliemann C et al (2007) Bevacizumab reduces
receptor-2 tyrosine kinase inhibitor for the treatment of VEGF expression in patients with relapsed and refractory
cancer. Cancer Res 65(10):4389–4400 acute myeloid leukemia without clinical antileukemic
Weidner N, Carroll PR et al (1993) Tumor angiogenesis activity. Leukemia 21(6):1310–1312
correlates with metastasis in invasive prostate carcinoma. Zhang W, D S, Yang D et al (2009) Genetic variants in
Am J Pathol 143(2):401–409 angiogenesis pathway associated with clinical outcome in
Weir HK, Thun MJ et al (2003) Annual report to the nation on NSCLC patients treated with bevacizumab in combination
the status of cancer, 1975–2000, featuring the uses of with carboplatin and paclitaxel: Subset pharmacogenetic
surveillance data for cancer prevention and control. J Natl analysis of ECOG 4599. J Clin Oncol 27:8032
Cancer Inst 95(17):1276–1299 Zhou ZT, Zhou FX et al (2011) Phase II study of cisplatin/
Werb Z, Vu TH et al (1999) Matrix-degrading proteases and etoposide and endostar for extensive-stage small-cell-lung
angiogenesis during development and tumor formation. cancer. Cancer Chemother Pharmacol (Epub ahead of
APMIS 107(1):11–18 print)
Interventional Pulmonology
Branislav Perin, Bojan Zarić, and Heinrich D. Becker

Contents Abstract
Interventional pulmonology is relatively new field
1 Introduction.............................................................. 46 within pulmonary medicine focused on use of
1.1 Diagnostic Interventional Pulmonology advanced bronchoscopy methods and interven-
Techniques ................................................................. 46
1.2 Therapeutic Interventional Pulmonology
tional techniques in diagnosis and therapy of
Techniques ................................................................. 48 respiratory diseases. Various respiratory disorders
may result in central airway obstruction (CAO),
References.......................................................................... 52
and central airway obstruction can cause signifi-
cant morbidity and mortality. Exact data on
incidence and prevalence of central airway
obstruction are not available, but epidemiological
studies investigating lung cancer are suggesting
that CAO is frequent and significant part of
morbidity and mortality in lung cancer patients.
These studies suggest that 20-30% of lung cancer
patients are experiencing complications due to
CAO, and that 40% of lethal outcomes are related
to CAO. The treatment of patients with CAO
requires not only understanding of etiology,
physiology, diagnostic and therapeutic procedures
but also availability of multidisciplinary team com-
posed of interventional pulmonologists, thoracic
surgeons, pulmonologists, oncologists, anesthesi-
ologists and radiologists. Evaluation of procedures
and their efficacy is extremely difficult; randomiza-
tion of studies in this field is extremely complicated.
In one hand it is difficult to find patients with
comparable disorder and comorbidity, on the other
B. Perin (&)  B. Zarić hand all of the patients are critically ill and random-
Institute for Pulmonary Diseases of Vojvodina, ization in this case is unethical. That is the reason why
Clinic for Pulmonary Oncology, Faculty of Medicine,
University of Novi Sad, Novi Sad, Serbia the majority of studies are retrospective analyses.
e-mail: bperin@eunet.rs One thing is for certain—the use of novel technology
H. D. Becker and interventional procedures leads to improvement
Department for Interdisciplinary Endoscopy, in survival and quality of life of patients with CAO.
Thoraxklinik, Heidelberg, Germany

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 45
DOI: 10.1007/174_2011_206, Ó Springer-Verlag Berlin Heidelberg 2011
46 B. Perin et al.

The primary question is no more ‘‘Is therapy electrocautery, argon plasma coagulation and place-
helpful?’’ but ‘‘Which therapy is best for particular ment of tracheobronchial stents. Other therapeutic
patient?’’. interventions can proceed, after the initial treatment
and re-opening of the airway. Interventional pulmon-
ology techniques with delayed effect can improve the
1 Introduction control of symptoms and quality of life (QoL), and
addition of specific chemoradiotherapy regimen can
In recent years interventional pulmonology tech- have certain impact on disease-free period and survival.
niques have found their place in the palliative treat- Every symptomatic malignant CAO must be con-
ment of lung cancer invading central airways (trachea sidered as a potentially fatal one. Adequate bron-
and principal bronchi). The curative effect of inter- choscopic evaluation can offer appropriate indication
ventional techniques is reported in number of studies for imminent interventional technique for rapid air-
with very different success ratio, but with excellent way desobstruction. Interventional pulmonology
potential and perspective. Increase in number and techniques are extremely expensive and they should
variety of these techniques led to the development of be available only to the top respiratory institutions.
internationally accepted guidelines for their use. Deployment of these techniques requires a multidis-
The choice of specific interventional technique in the ciplinary team of interventional pulmonologists,
treatment of lung cancer patients with central airway anesthesiologists, thoracic surgeons, radiologists and
stenosis (CAO) depends on several factors: patient’s oncologists. Institutions where interventional tech-
general condition and co-morbidities, type and niques are implemented need to have appropriate
characteristics of airway stenosis and availability of facilities, well-trained personnel and a variety of
techniques and trained personnel. interventional techniques available at any time
Interventional techniques can be divided into diag- (Colt, Murgu 2010; Sutedja 2003; Wahidi et al 2007).
nostic and therapeutical. Diagnostic interventional
techniques, among others are: autofluorescence bron-
choscopy (AFB), endobronchial ultrasound (EBUS), 1.1 Diagnostic Interventional
electromagnetic navigation bronchoscopy and narrow Pulmonology Techniques
band imaging bronchoscopy. Therapeutic techniques
are divided into ones with imminent desobstruction of 1.1.1 Autofluorescence Videobronchoscopy
central airways and ones with delayed effect. Thera- Autofluorescence imaging videobronchoscopy (AFI) is
peutical interventional pulmonology techniques can be one of the new systems of autofluorescence bronchos-
curative or palliative. In cases of carcinoma in situ or copy designed for thorough examination of bronchial
early invasive lung cancer, interventional pulmonology mucosa. The integration of autofluorescence and vid-
can offer a variety of techniques with curative effect. eobronchoscopy provides clear images of normal and
Photodynamic therapy (PDT), cryotherapy, electro- pathologically altered bronchial mucosa. Major indi-
cautery (EC), argon plasma coagulation (APC) and cations for AFI include evaluation of early-stage
brachytherapy are reported to have curative potential, lung cancer and detection of pre-cancerous lesions.
in cases of early-stage lung cancer. Bronchoscopic However, in recent years the indications for AFI are
treatment of early-stage lung cancer requires precise widening and this tool might find its place in daily
and accurate staging of the disease, using all available routine bronchoscopic practice. With new indications
advanced techniques for staging, e.g., positron emis- for AFI, such as evaluation of tumor extension or fol-
sion tomography, endobronchial ultrasound and auto- low-up after surgical resection, this tool might be more
fluorescence or narrow-band imaging bronchoscopy to often used by bronchoscopists. Sharp learning curve
make a distinction between non-invasive and invasive and clear designation between healthy and pathologi-
bronchial carcinoma and to determine nodal status with cally altered mucosa make this technology acceptable
reliable accuracy. for young and inexperienced bronchoscopists.
Palliative interventional pulmonology techniques One of the major disadvantages of AFI is low
are aimed at relieving of symptoms of malignant CAO. specificity in detection of pre-malignant lesions and
Techniques with imminent effect are laser resection, early-stage lung cancer. This disadvantage could be
Interventional Pulmonology 47

overcome with the appearance of new and improved wave length ranging from 390 to 445 nm, for imaging
technologies in autofluorescence such as: addition of the capillaries of the surface mucosal layer. A green
backscattered light analysis, ultraviolet spectra, fluo- narrow band (of the wave length from 530 to 550 nm)
rescence-reflectance or dual digital systems. Quanti- is used to visualize the thick blood vessels inside the
tative image analysis is also one of the ways to improve membranes. This approach provides a better contrast
objectivity and minimize observer errors. However, on the mucosal surface, reduces examination time and
one of the most appropriate solutions would be addition eliminates futile biopsies.
of AFI to narrow-band imaging (NBI), and merging of The use of the NBI technology in the detection of
two technologies into one videobronchoscope. New lung cancer started with the development of magni-
systems for autofluorescence, such as autofluorescence fying videobronchoscopy and integration of these two
imaging–AFI (Olympus Co, Tokyo, Japan), SAFE- systems. Most recent publications confirm that NBI
3,000 (Pentax Co, Japan), D-Light AF system enables better visualization of the bronchial mucosa
(Karl Storz, Tuttlingen, Germany) or Onco-LIFE and differentiation between malignant and non-
(Xillix Technologies Corp. Richmond, Canada), pro- malignant tissue. NBI proves to be more efficient in
duce clear, high-quality images of bronchial mucosa. the detection of pre-cancerous lesions, especially
Images generated by the new systems are easy to inter- angiogenic squamous dysplasia (ASD), than the white
pret and characteristic. In the mentioned systems auto- light videobronchoscopy alone. The learning curve
fluorescence and white light videobronchoscopy are for NBI videobronchoscopy is sharp, and that fact
incorporated into one scope, making it easy to switch makes the NBI useful even when used by an inex-
between modes of examination. On the AFI examination perienced bronchoscopist. It is believed that the
normal mucosa appears green, while pathologically combination of NBI with autofluorescence video-
altered mucosa is magenta or red-brownish (Yarmus, bronchoscopy will give even better results in lung
Feller-Kopman 2010; Yasufuku 2010). cancer detection.

1.1.2 Narrow-Band Imaging 1.1.3 Endobronchial Ultrasound


Narrow-band imaging (NBI) is a new endoscopic There are currently two major types of endobron-
technique designed for detection of pathologically chial ultrasound (EBUS), linear and radial. Linear
altered sub-mucosal and mucosal microvascular EBUS is commonly used for mediastinal staging of
patterns. The combination of magnification video- lung cancer, or real-time TBNA (transbronchial
bronchoscopy and NBI showed great potential in needle aspiration). This approach enables the bron-
detection of pre-cancerous and cancerous lesions of the choscopist to visualize lymph nodes even smaller
bronchial mucosa. The preliminary studies confirmed than 1 cm in diameter, allowing accurate nodal
supremacy of NBI over white light videobronchoscopy staging. The technique is proven to be completely
in the detection of pre-malignant and malignant complementary with mediastinoscopy. On the other
lesions. Pathological patterns of capillaries in bron- hand real-time EBUS guided TBNA allows more
chial mucosa are known as Shibuya’s descriptors comfortable mediastinal re-staging which is neces-
(dotted, tortuous and abrupt ending blood vessels). sary after neoadjuvant radiochemotherapy. Radial
When respiratory endoscopy is concerned, the NBI is endobronchial probe allows insertion of the probe
still a ‘‘technology in search of proper indication’’. through the working channel of the diagnostic
More randomized trials are necessary to confirm the bronchoscope. It can also be used for mediastinal
place of NBI in the diagnostic algorithm, and more staging; however, this technique enables excellent
trials are needed to evaluate the relation of NBI to insight into the pulmonary parenchyma. Radial
autofluorescence videobronchoscopy, and white light EBUS is routinely used for diagnosis of peripheral
magnification videobronchoscopy. Considering the lung cancers, as well as for the assessment of tumor
fact that NBI examination of the tracheo-bronchial tree penetration into the bronchial wall. Considering the
is easy, reproducible and clear to interpret, it is certain fact that up to nine layers of bronchial wall can be
that the NBI videobronchoscopy will play a significant visualized by radial EBUS, this technique is irre-
role in the future of lung cancer detection and staging. placeable for the assessment of bronchial wall in
The NBI system uses a blue narrow band, with the early-stage lung cancer.
48 B. Perin et al.

Fig. 1 White light videobronchoscopy image of the tumor in Fig. 2 Autofluorescence imaging (AFI) videobronchoscopy
the distal part of intermediary bronchus image of the tumor in the distal part of intermediary bronchus

Comparative images of white light videobron- tissues. The result is vaporization, coagulation and
choscopy, AFI, NBI and radial EBUS on the same necrosis of the targeted tissue. There are several types
lesion are presented in Figs. 1, 2, 3, 4. of lasers used in practice (Nd:YAG, CO2, argon, dye,
diode, and YAP:Nd), only Nd:YAG (neodym-
1.1.4 Novel Diagnostic Techniques ium:yttrium aluminium garnet) is widely used in
in Interventional Pulmonology pulmonology. The effects of laser beam depend on
There are several newly developed techniques that several factors, among others, power density,
entered the arena of diagnostic interventional pul- absorption and scattering ratio of soft tissues and
monology in the last years. Electromagnetic naviga- delivery system but one of the most important factors
tion (EMN) bronchoscopy is successfully deployed as determining biological effect of laser on soft tissues is
a guiding tool for diagnosis of peripheral lung cancer. the wavelength of the laser light. Wavelength of laser
In recent years other techniques are available: determines the absorption and through the effect of
enhanced bronchoscopic navigation, confocal fluo- delivered heat energy on the tissue. The wavelength
rescence microscopy (endoscopy), optical coherence of Nd:YAG laser is 1,064 nm, it is in invisible range
tomography and ultrathin bronchoscopy (Yarmus, of infrared region and needs a pilot light (usually red)
Feller-Kopman 2010; Yasufuku 2010). for its guidance.
Major indication for laser photoresection is
malignant or non-malignant central airway obstruc-
1.2 Therapeutic Interventional tion due to intraluminal growth of malignant or
Pulmonology Techniques benign tissue. Lesions most suitable for laser resec-
tions are situated centrally (trachea and mainstream
1.2.1 Techniques with Imminent Effect bronchi), short in length (B4 cm), with visible distal
bronchial lumen and functional lung distal to the
1.2.1.1 Nd:YAG Laser Photoresection obstruction.
‘‘Light amplification of stimulated emission of Nd:YAG laser re-section can be carried out via
radiation’’ or LASER is one of the most explored flexible bronchoscopy or the combination of rigid and
principles in interventional pulmonology. Laser flexible bronchoscopy under general anesthesia. The
beam delivers energy in form of the heat to the target initial power setting is usually about 40 W, with pulse
Interventional Pulmonology 49

lesion is advisable. Laser re-section is usually per-


formed under general anesthesia, as a precaution
oxygen concentration should be kept under 40% in
order to prevent airway fire.
Absolute contraindication is extraluminal disease;
relative contraindications include recent myocardial
infarction, ventricular arrhythmias, conduction
abnormalities, hypotension, decompensated heart
failure, severe obstructive lung disease, extensive
tumor involvement, unresolved coagulopathies and
sepsis.
The overall complication rate of Nd:YAG laser
resection is generally low. Possible complications
include hypoxemia intraoperatively and post-opera-
tively, hemorrhage, airway perforation, airway fire
(burns), pneumothorax and fistulae formation. After
the treatment patient should be observed in the
recovery room for a reasonable period of time
because of the possibility of bronchospasm or
Fig. 3 Narrow band imaging videobronchoscopy image of the
tumor in the distal part of intermediary bronchus
laryngospasm. With adequate precaution measures
taken complication rate is usually less than 5%
(Colt, Murgu 2010; Sutedja 2003; Wahidi et al
2007).

1.2.1.2 Electrocautery
Electrocautery (EC) represents a contact form of
electrosurgery. Electron flow between the tip of the
probe and the tissue is a result of voltage difference
between these two surfaces. Electrons are then
transmitted through a tiny gap of air between these
two surfaces. Created electrical current is affecting
target tissue in the form of heat, causing coagulation,
carbonization or vaporization of the tissue. The cur-
rent leaves the body through a grounding plate, usu-
ally applied on patient’s arm. The effects of EC
depend on power setting, tissue resistance, time of
application and the applied mode of EC. Low voltage,
Fig. 4 Linear endobronchial ultrasound (EBUS) image of the low power and high amperage will cause coagulation.
tumor in the distal part of intermediary bronchus High voltage, high power and low amperage will
cause carbonization. There is ‘‘cut’’ mode and
‘‘blended’’ mode of EC, the last one is preferred for
duration of 0.5–1 s. The tip of the probe is aimed its ability to combine cutting and coagulation. Some
1 cm proximally and parallel to the lesion. Tissue authors divide EC coagulation in three types; soft—to
effect of Nd:YAG laser can be adjusted either by avoid carbonization, hard—for deeper tissue pene-
increasing the power setting or by moving the tip of tration and spray—for surface haemostasis. Indica-
the probe further or closer to the target lesion. The tions for EC are the same as for laser re-section,
result will be vaporization, coagulation or necrosis of and often EC is used as a cheaper alternative to laser
the tissue. The depth of penetration of laser beam is re-section. Intraluminal obstruction of major airways
not immediately visible, so frequent re-analysis of the due to benign or malignant tissue proliferation is most
50 B. Perin et al.

common indication for its use. Absolute contraindi- The effect of APC depends on power setting,
cations are extraluminal disease and a pacemaker application time and conductivity of the tissue.
susceptible to electrical interference. Coagulated tissues have higher resistance and higher
Electrocautery can be carried out via rigid or resistance lowers conductivity and limits penetration.
flexible bronchoscopy, or the combination of these At power setting of 40–120 W and burst time less
two techniques. When rigid bronchoscopy is per- than 2 s, the penetration depth is limited to less than
formed EC is performed under general anesthesia. 5 mm. One of major benefits of APC is the fact that
Analgosedation modality can be used in flexible the lesions situated laterally to the probe or the lesions
bronchoscopy EC. A specially isolated ceramic-tip ‘‘around the corner’’ can be successfully treated, in
flexible bronchoscope is used for EC, and the patient contrast to laser or electrocautery. Disadvantages of
is electrically grounded with a pad or a plate. Limiting APC include low penetration ability; therefore the
inhaled oxygen fraction and avoiding flammable treatment of bulky tumor masses APC is not recom-
materials (plastic endotracheal tube or tracheobron- mendable. In that case other interventional techniques
chial stents) are necessary precaution measures. There usually must follow APC. Repeated bronchoscopic
are several types of EC probes designed to achieve checkups are also usually required in order to remove
wanted effects on target tissue. Blunt probe is usually all the necrotic debris from the airways.
used for coagulation and carbonization, knife causes Indications for APC are similar to EC or laser
coagulation and blend, and snare is designed for resections. They include intrinsic airway obstruction
blend. Coagulative effect of EC significantly corre- due proliferation of malignant or benign tissue. Most
lates with histologic tissue damage effect on the tar- suitable lesions have large endobronchial component
geted tissue. with visible distal bronchial lumen and functional
Possible complications of EC include airway per- pulmonary tissue. The major indication for APC is
foration, electrical shocks and hemorrhage, however, hamostasis in hemoptysis. Since APC affects wide
only mild hemorrhage has been reported in published surface it is a recommendable technique for hem-
studies. Precaution measures for application of EC optysis control. APC is successfully used in re-section
include limited power setting (40 W), low inspired of granulation tissue proliferating through the pores of
oxygen concentration (B40%), short burst time metallic stent; there are also reports of use in the
(B2 s) and the use of isolated bronchoscope. EC treatment of respiratory papillomatosis and post-
seems to be good alternative to laser resection; it is transplantation benign tracheobronchial stenosis.
cheap and safe technique for urgent airway debulking. There are no absolute contraindications for APC use,
except for extraluminal disease. Relative contraindi-
1.2.1.3 Argon Plasma Coagulation cations are the same as for laser re-sections and EC.
Argon plasma coagulation (APC) is a form of non- APC can be carried out via flexible bronchoscopy
contact electrosurgery that uses ionized argon gas alone; however, the combination of flexible and rigid
transformed into plasma in order to create electrical bronchoscopy allows better control of bursts and
current. When high voltage spark (5,000–6,000 V) adequate removal of debris during the intervention.
ignites argon gas, the gas is ionized into plasma. The technique is usually performed under general
Monopolar current of ionized plasma affects target anesthesia and requires all the necessary precautions
tissue in the form of heat, producing coagulative and for safe general anesthesia. Oxygen concentration
necrotic effect on target tissue. Plasma seeks the way should be under 40% and all flammable materials e.g.,
of least resistance, targeting directly wide surface of silicone stents or endotracheal tube, should be avoi-
tissue. Closed current circuit is necessary for argon ded. Initial settings for application of APC include:
plasma to flow; the tip of the probe must therefore be power at 30–80 W, burst time of 2–3 s and argon gas
situated less than a centimeter from the tissue. If the flow 0.3–2 L/min. The flexible probes for APC are
distance between the tip of the probe and the tissue is 1.5 or 2.3 mm in diameter and usually 200 cm long.
more than 1 cm, the circuit will be open and the effect Probes can easily pass through working channel of
will be lost. The current leaves the body through a flexible bronchoscope. The tip of the probe must
grounding plate usually situated under patient’s lower protrude at least 1 cm off the tip of the bronchoscope.
back. In makes the visualization field clear and prevents
Interventional Pulmonology 51

possible burning of the bronchoscope. The probe must of the airway, serious bleeding and trauma. Confir-
be kept 1 cm from the target tissue in order to keep mation of stent position requires fluoroscopy; tissue
the flow of plasma. During the procedure the debris (benign or malignant) grows through the stent
should be removed with forceps or with suction. requiring additional re-sections and these stents are
Possible complications include airway perforation expensive. Most suitable stents at the current moment
(pneumomediastinum or pneumothorax), airway fire are hybrid ones, metallic fully covered stents. The
and damage to the bronchoscope. Incidence of these best solution for the treatment of obstruction with
complications is less than 1%. Limitation of oxygen carinal involvement is Y shaped dynamic stent. The
concentration on less than 40%, keeping power set- dynamic stent is a hybrid stent made of silicone with
tings under 80 W with application time less than 5 s, steel claps.
minimizes the probability of these complications. One of techniques for urgent airway debulking
In conclusion, APC is recommendable technique for usually precedes stent insertion in malignant and
the management of hemoptysis and for removing of benign central airway obstruction (CAO). With laser
CAO. resection, EC or APC intraluminal component can
be easily removed and stent application can follow.
1.2.1.4 Tracheobronchial Stents In such situation one usually decides to place silicone
Tracheobronchial stents have four major indications or hybrid stent.
in interventional pulmonology; (1) extrinsic com- The ideal stent of the future should be easy to insert
pression from tumors or enlarged lymphnodes, (2) and remove, with low possibility of migration, rigid
stabilizing airway patency after removal of endo- enough to support the airway and flexible enough for
bronchial malignancy, (3) sealing of tracheo-oesoph- adaptation to airway structure. It should be biologi-
ageal fistulas and (4) treatment of benign central cally inert to minimize granulation tissue formation,
airway obstruction. Tracheobronchial stents are divi- available in different sizes and not too expensive.
ded into two large groups; silicone and metallic There are some feasibility studies going on, in order to
stents. Metallic stents can be balloon expandable and investigate the successfulness of tracheobronchial
self-expanding. Baloon expandable metallic stents are stents made of bioabsorbable materials.
currently out of date, and rarely used. Both types of
stents have certain advantages and disadvantages, 1.2.2 Techniques with Delayed Effect
also, both types of stents have specific indications and
complications. Commonly used in these days are 1.2.2.1 Bronchoscopic Balloon Dilatation
hybrid stents, made of metal wire mesh and covered Balloon dilatation (bronchoplasty) can be successfully
with silicone (UltraflexÒ [Boston scientific] and Aero used for the treatment of malignant and non-malignant
stentÒ [Alveolus]). Silicone stents are adjustable to central airway obstruction. This technique is success-
the airway architecture, easily removed, with no in fully used for the treatment of post-intubation or post-
growth of granulation or tumor tissue, non-reactive in transplantation stenoses or as an introductory technique
the means of mucosa irritation, their expansion is in the treatment of malignant obstruction before stent-
easily controlled and they are cheaper than metallic ing. Standard procedure requires assessment of the
stents. However, placement of silicone stents requires endoscopic lesion, with detection of its proximal and
rigid bronchoscopy under general anesthesia, the distal ending. Guide-wire is then inserted through the
placement is rather complicated, and one of most working channel of the bronchoscope and placed into
common problems is dislocation. Some authors the region of the obstruction. Balloon catheter is placed
complain that the decreased inner diameter leads to through the guide-wire under fluoroscopy control.
impaired mucus clearance and formation of mucus Balloon inflation and deflation is followed endoscopi-
plugs. Metallic stents are easily delivered via flexible cally and fluoroscopically, the dilatation can be
bronchoscope, stable in place and the position of the repeated several times. Major indications include
stent is confirmed on fluoroscopy. On the other hand, the preparation of the stenotic bronchial segment for
once placed these stents are permanent, the position the implantation of the stent or brachytherapy
of the stent can not be adjusted and removal of the catheter. Possible complications include perforation of
stents is dangerous because it may cause a perforation the bronchi or re-stenosis.
52 B. Perin et al.

1.2.2.2 Endobronchial Brachytherapy late complications of HDR EBBT. While early studies
Endobronchial brachytherapy (EBBT) allows appli- showed small percentage of complications, latter
cation of maximal radiotherapy dose directly to tumor studies revealed more frequent complications, espe-
with minimal damage for surrounding tissue. cially hemoptysis. Usually reported complication rate
High-dose rate endobronchial brachytherapy is less than 5%, however, in some studies complica-
(HDR-EBRT) is one of the commonly used interven- tions appeared at the rate of 35%.
tional pulmonology techniques with delayed effect in
the treatment of malignant endobronchial obstruction. 1.2.2.3 Other Techniques
This technique found its place in all of the guidelines Cryotherapy is one of the techniques for debulking of
for the use of interventional pulmonology in lung central airways. It is based on application of extre-
cancer. Major number of studies reported influence of mely low temperatures (under -40°C) in order to
HDR EBBT on the time to progression in advanced freeze and destroy malignant tissue. The effect of
lung cancer treatment. As a part of multi-modality cryotherapy depends on the speed of freezing and
treatment of advanced lung cancer, HDR EBBT might thawing, lowest temperature, number of freeze thaw
influence survival in patients with malignant central cycles and cellular amount of water. Freezing under
airway obstruction. High-dose rate EBBT can be used -40°C with speed of 100°C/min experimentally
as a sole technique for desobstruction, in non-urgent destroys 90% of the cells.
cases of bronchial obstruction. In cases where bron- Photodynamic therapy represents activation of
chial obstruction is urgent, EBBT can safely follow photosensitive substance with non-thermal laser light.
laser resection, electrocautery or argon plasma coag- This results in phototoxic reaction, which leads to
ulation. Palliative effect of EBBT is recognized in cellular death. This technique is suitable for the
majority of the studies that investigated its effect on treatment of early-stage superficial lung cancer.
dyspnea, cough, post obstructive pneumonia, hem-
optysis and other symptoms of lung cancer. In recent
studies HDR EBBT was used for treatment of early- References
stage lung cancer and benign central airway obstruc-
tion. HDR EBBT was successfully combined with Colt HG, Murgu SD (2010) Interventional bronchoscopy from
photodynamic therapy in the treatment of bulky lung bench to bedside: new techniques for early lung cancer
detection. Clin Chest Med 31(1):29–37
cancer. In the future HDR EBBT in combination with Sutedja G (2003) New techniques for early detection of lung
electromagnetically navigated bronchoscopy could cancer. Eur Respir J 21(39):57–66
find its place in the treatment of inoperable peripheral Wahidi MM, Herth FJF, Ernst A (2007) State of the art:
lung cancer. However, precise protocols for HDR interventional pulmonology. Chest 131:261–274
Yarmus L, Feller-Kopman D (2010) Bronchoscopes of the
EBBT do not exist. Still, there are major differences in twenty-first century. Clin Chest Med 31(1):19–27
timing of fractions and total dose. Therefore, there are Yasufuku K (2010) Early diagnosis of lung cancer. Clin Chest
significant differences in the appearance of early and Med 31(1):39–47
Pathology of Lung Cancer
Mary Beth Beasley

Contents Abstract
Over the past decade, the diagnosis, treatment
1 Introduction.............................................................. 53 and management of lung cancer has evolved
2 Adenocarcinoma ...................................................... 54 substantially, in large part to the development of
2.1 Background ................................................................ 54 novel chemotherapeutic agents and targeted
2.2 Bronchioloalveolar Carcinoma, Adenocarcinoma therapy in particular. As such, accurate classifi-
in situ and Minimally Invasive Adenocarcinoma .... 55
cation of lung carcinomas from a histologic
2.3 ‘‘Mixed Subtype’’ Adenocarcinoma ......................... 56
2.4 Mucinous Adenocarcinomas ..................................... 57 standpoint has become increasingly critical for
2.5 Adenocarcinoma Conclusions and Areas in Need appropriate patient management. Much of the
of Further Study ........................................................ 58 focus of lung cancer pathology in the past
3 Squamous Cell Carcinoma ..................................... 58 decade has concentrated on adenocarcinoma.
As such, the aim of this chapter will primarily
4 Molecular Testing in Lung Cancer ....................... 59
focus on the evolution of this cancer subtype and
5 Pathology of Lung Carcinomas Treated With review the recently published adenocarcinoma
Radiation and/or Neo-adjuvant Therapy ............. 60
classification prepared jointly by the International
References.......................................................................... 60 Association for the Study of Lung Cancer, the
American Thoracic Society and the European
Respiratory Society. Squamous cell carcinoma
will also be discussed, particularly in regard to
accurate discrimination from adenocarcinoma.
The role of the pathologist in molecular testing will
be reviewed, particularly as it relates to specimens
obtained from radiologic procedures. Finally, given
that the scope of this publication is radiation
oncology, the histologic changes and evaluation
of tumors which have undergone radiation and/or
neo-adjuvant therapy will be discussed.

1 Introduction
M. B. Beasley (&)
Mount Sinai Medical Center, Multiple subtypes of lung carcinoma have always
One Gustave L. Levy Place,
New York, NY 10128, USA existed from a pathologic standpoint, and the current
e-mail: mbbeasleymd@yahoo.com World Health Organization (WHO) classification is

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 53
DOI: 10.1007/174_2011_304, Ó Springer-Verlag Berlin Heidelberg 2011
54 M. B. Beasley

Table 1 2004 World Health Organization classification of carcinoma, with clinicians primarily being concerned
pulmonary malignant epithelial tumors (Travis et al. 2004) with whether a carcinoma was small-cell carcinoma
Pre-invasive lesions (SCLC) or non-small-cell carcinoma (NSCLC), the
Squamous cell carcinoma in situ later typically comprising squamous cell carcinoma
Atypical adenomatous hyperplasia (SCC), adenocarcinoma (ADC) and large cell carci-
Diffuse idiopathic neuroendocrine cell hyperplasia
noma. This clinical grouping has traditionally existed
as treatment for SCLC differed from that of NSCLC,
Squamous cell carcinoma
but did not differ significantly among the subtypes of
Variants-papillary, clear cell, small cell, basaloid
NSCLC (Travis et al. 2004; Beasley et al. 2005;
Adenocarcinoma Lim et al. 2008). The advent of expanded chemo-
Adenocarcinoma, mixed type therapeutic options and targeted therapies has shifted
Acinar adenocarcinoma this paradigm and placed a greater role on the
Papillary adenocarcinoma pathologist in regard to accurate subtyping of tumors
Bronchioloalveolar carcinoma as well as molecular testing (Pirker et al. 2010; Travis
Non-mucinous
et al. 2011). Clearly, the variety of malignant lung
tumors is myriad; however, the scope of this article
Mucinous
will focus on the evolving issues involving the most
Mixed
commonly encountered tumors, namely adenocarci-
Solid adenocarcinoma with mucin production noma and squamous cell carcinoma, with particular
Variants focus on adenocarcinoma. Practical issues regarding
Fetal adenocarcinoma tumor diagnosis and molecular testing as they pertain
Mucinous ‘‘colloid’’ carcinoma to specimens obtained from interventional radiolo-
Mucinous cystadenocarcinoma gists will also be discussed. The chapter will end with
Signet ring cell adenocarcinoma a brief discussion of the histologic features encoun-
tered in tumors which have undergone pre-operative
Clear cell adenocarcinoma
or neo-adjuvant therapy.
Small-cell carcinoma
Combined small-cell carcinoma
Large-cell carcinoma 2 Adenocarcinoma
Large-cell neuroendocrine carcinoma
Combined large-cell neuroendocrine carcinoma 2.1 Background
Other variants-basaloid carcinoma, lymphoepithelioma-like
carcinoma, clear cell carcinoma, large-cell carcinoma with Adenocarcinoma now represents the most common
rhabdoid phenotype subtype of lung carcinoma. The current WHO clas-
Adenosquamous carcinoma sification divides adenocarcinoma into acinar, papil-
Sarcomatoid carcinoma lary, bronchioloalveolar and solid types, along with
Variants-pleomorphic carcinoma, spindle cell carcinoma,
several other variants including mucinous carcino-
giant cell carcinoma, carcinosarcoma and pulmonary mas. The WHO classification also includes a category
blastoma of ‘‘mixed type adenocarcinoma’’ which, given the
Carcinoid tumor known histologic heterogeneity of lung carcinomas,
Typical carcinoid encompasses up to 90% of all lung adenocarcinomas
Atypical carcinoid and thus provides minimal information of clinical
relevance (Travis et al. 2004). As such, a new clas-
Salivary gland tumors—mucoepidermoid carcinoma, adenoid
cystic carcinoma, epithelial-myoepithelial carcinoma sification of adenocarcinoma was recently proposed
jointly by the International Association for the Study
of Lung Cancer (IASLC), the American Thoracic
presented in Table 1 (Travis et al. 2004). In spite Society (ATS) and the European Respiratory Society
of this, the role of the pathologist has traditionally (ERS). This new classification scheme, which will be
been relatively limited in the management of lung referred to as the ‘‘IASLC classification’’ in this text,
Pathology of Lung Cancer 55

Table 2 IASLC/ATS/ERS adenocarcinoma classification


(Travis et al. 2011)
Pre-invasive lesions
Atypical adenomatous hyperplasia
Adenocarcinoma in situ (B3.0 cm, formerly
bronchioloalveolar carcinoma)
Non-mucinous
Mucinous
Mixed mucinous/non-mucinous
Minimally invasive adenocarcinoma (B3.0 cm lepidic
predominant tumor with B5 mm of invasion)
Non-mucinous
Mucinous
Mixed mucinous/non-mucinous Fig. 1 Adenocarcinoma with non-mucinous lepidic growth
(H and E 2009). Non-mucinous tumor cells grow along pre-
Invasive adenocarcinoma
existing alveolar septa. Tumors consisting purely of this growth
Lepidic predominant (formerly non-mucinous BAC pattern pattern are classified as adenocarcinoma in situ in the IASLC
with [5 mm of invasion) classification system
Acinar predominant
Papillary predominant
Micropapillary predominant into non-mucinous and mucinous subtypes (Travis
Solid predominant with mucin production
et al. 2004). The term ‘‘lepidic’’ refers to growth
along pre-existing alveolar walls and derives from the
Variants
greek word ‘‘lepis’’ meaning ‘‘scales’’. As strictly
Invasive mucinous adenocarcinoma (formerly mucinous
bronchioloalveolar carcinoma)
defined, isolated non-mucinous BAC generally
appears as a ground glass lesion radiographically and
Colloid carcinoma
is associated with a nearly 100% 5-year survival, as
Fetal (low and high grade)
first noted by Noguchi et al. (1995) and subsequently
Enteric supported by other studies (Suzuki et al. 2002,
Sakurai et al. 2004a; Borczuk et al. 2009). The BAC
is presented in Table 2 (Travis et al. 2011). The pri- terminology has unfortunately been inappropriately
mary issues addressed by the new classification which applied by pathologists and clinicians alike, which has
differ significantly from the WHO classification are led to much confusion in the literature. In an attempt
the issue of bronchioloalveolar carcinoma (BAC) and to emphasize the strict definition and prognostic
the heterogeneous nature of the ‘‘mixed subtype’’ of implications, the IASLC classification has proposed
adenocarcinoma, and the issue of mucinous carcino- that the term ‘‘adenocarcinoma in situ (AIS)’’ be used
mas of the lung. The classification also emphasizes for solitary lesions with pure lepidic growth (Fig. 1).
the need to accurately discriminate between ADC and The IASLC classification retains the current WHO
SCC and the importance of appropriate tissue man- definition for BAC but additionally emphasizes that
agement for molecular testing. papillary and micropapillary growth patterns, as well
as significant intra-alveolar tumor cells should be
2.2 Bronchioloalveolar Carcinoma, absent (Travis et al. 2011).
Adenocarcinoma in situ A second significant change in the IASLC classi-
and Minimally Invasive fication is the addition of a category of ‘‘minimally
Adenocarcinoma invasive adenocarcinoma (MIA)’’. Several studies
have evaluated the impact on survival of the amount
In the WHO classification, BAC is defined as an of stromal invasion present in a tumor otherwise
adenocarcinoma with pure lepidic growth without showing non-mucinous lepidic growth (Suzuki et al.
stromal, pleural or vascular invasion, and is divided 2002; Sakurai et al. 2004a, b; Yim et al. 2007;
56 M. B. Beasley

Borczuk et al. 2009). Based on these studies, tumors


with 5 mm of invasion or less have the same excellent
prognosis as those with pure lepidic growth. As such,
the newly proposed category of MIA applies to
tumors with 5 mm or less of invasion in any one
focus, with the invasive component defined as any
histologic subtype other than lepidic growth or any
tumor cells infiltrating a fibroblastic stroma. The term
MIA is not used if the tumor invades lymphatics,
blood vessels or pleura, or if tumor necrosis is pres-
ent, even if the invasive focus is \5 mm overall
(Travis et al. 2011).
A cautionary note in regard to both AIS and MIA
is that both designations apply to tumors which are
localized and 3.0 cm or less in size. This is primarily Fig. 2 Pulmonary adenocarcinoma with papillary morphol-
ogy. Note the branching papillary structures with fibrovascular
because the majority of the research on these tumors cores (H and E 2009)
has been done on stage 1 adenocarcinomas and the
implications in larger tumors are as of yet uncertain.
Additionally, both diagnoses require that the entire
tumor be histologically completely evaluated, and
thus these designations should be made only on
resected specimens and are not appropriate diagnoses
for small biopsy or cytology specimens. Further, both
AIS and MIA refer predominantly to non-mucinous
tumors (Travis et al. 2011). While a mucinous sub-
type of BAC is present in the WHO classification and
is retained in the IASLC classification of AIS and
MIA, mucinous tumors meeting the strict criteria are
exceedingly rare and almost always contain signifi-
cant foci of invasion (Travis et al. 2004, 2011).

Fig. 3 Pulmonary adenocarcinoma with micropapillary mor-


2.3 ‘‘Mixed Subtype’’ Adenocarcinoma phology, characterized by small free-floating tufts of tumor
surrounding a fibrotic core (H and E 4009)
Using the current WHO classification, up to 90% of
all lung ADC fall into the category of ‘‘mixed sub-
type’’ (Beasley et al. 2005). As one would expect, this The IASLC classification emphasizes that papillary
encompasses a markedly heterogeneous group of and micropapillary growth are forms of invasive
tumors of differing clinical significance. In an attempt carcinoma, which is not always intuitive from a his-
to address this issue, the IASLC classification rec- tologic standpoint as these growth patterns may show
ommends that tumors with multiple growth patterns extensive intra-alveolar involvement rather than
should be classified by the predominant growth pat- conventional stromal invasion (Travis et al. 2011).
tern, and further recommends that any additional Papillary carcinoma is defined as carcinoma forming
patterns be indicated in 5% increments. In addition to true papillae with a fibrovascular core (Fig. 2). The
the lepidic growth pattern discussed above, the micropapillary pattern, in contrast, consists of small
IASLC classification retains acinar, papillary and tufts of tumor radiating from a central fibrotic core
solid patterns as forms of invasive carcinoma, and (Fig. 3). This subtype of adenocarcinoma has been
formally recognizes the micropapillary pattern which associated with aggressive behavior and an increased
was not included in the current WHO classification. incidence of lymph node metastases in stage 1 tumors
Pathology of Lung Cancer 57

(Amin et al. 2002; Miyoshi et al. 2003; Hoshi et al.


2004; Makimoto et al. 2005; Kuroda et al. 2006;
Kawakami et al. 2007; Kamiya et al. 2008; Borczuk
2009).
Histologic growth patterns have been associated
with behavior and to a certain degree with certain types
of mutations. Predominant solid and micropapillary
growth are generally associated with more aggressive
behavior, predominant lepidic growth with a more
favorable prognosis, and acinar and papillary growth
with an intermediate prognosis (Travis et al. 2011). In
regard to mutational status, epidermal growth factor
receptor (EGFR) mutations are found more commonly
in tumors with lepidic or papillary growth whereas
mucinous tumors frequently harbor KRAS mutations
and lack EGFR mutations (Dacic 2008, Dacic et al.
2010; Sartori et al. 2009; Hata et al. 2010) Solid tumors
with signet ring morphology have been correlated with
the presence of EML4-ALK translocations (Rodig et al.
2009). Therefore, it is hoped that the more detailed
information provided in the IASLC classification will
provide more meaningful information to the clinician
in regard to prognosis and potential management.

2.4 Mucinous Adenocarcinomas

In the past decade it has become increasingly clear Fig. 4 Invasive mucinous carcinoma characterized by areas of
lepidic growth along pre-existing alveolar septa (a, H and E
that mucinous carcinomas differ markedly from their 4009) and invasive (b, H and E 2009) growth
non-mucinous counterparts, not only morphologi-
cally, but also on immunohistochemical and molec-
ular grounds (Copin et al. 2001; Rossi et al. 2004; Signet ring carcinoma is not included as a formal
Brownlee et al. 2005; Finberg et al. 2007; Pirker et al. category, as this is felt to be primarily a cytologic
2010; Travis et al. 2011). As previously stated, the change and it is unclear if it is independently corre-
vast majority of tumors previously classified as lated with a worse prognosis or if the association is
mucinous BAC contains areas of invasion and it is due to the fact that signet ring cells are more often
extremely rare to encounter a mucinous tumor with associated with solid growth, which itself has a poor
lepidic growth that would meet the strict criteria for prognosis (Travis et al. 2011). It is recommended that
AIS or MIA (Fig. 4a, b). The IASLC classification this feature be included as a descriptor, however,
includes the categories of invasive mucinous carci- given its association with EML4-ALK translocations
noma (which would encompass most tumors previ- (Rodig et al. 2009). Finally, enteric adenocarcinoma
ously classified as mucinous BAC) and colloid has been included as a formal category. These tumors
carcinoma (characterized by dissecting pools of have morphologic and immunohistochemical overlap
extracellular mucin containing free-floating tumor). with gastrointestinal and colorectal carcinomas,
The uncommon fetal adenocarcinoma, in which which is problematic for the pathologist if there is an
glandular components resemble those of embryonic issue of whether a tumor is primary or metastatic.
lung, is retained as well, while mucinous cystadeno- Whether these tumors have unique clinical or
carcinoma has been dropped as such tumors are molecular features is currently uncertain (Rossi et al.
generally felt to be variants of colloid carcinoma. 2004; Inamura et al. 2005; Travis et al. 2011).
58 M. B. Beasley

2.5 Adenocarcinoma Conclusions


and Areas in Need of Further Study

In summary, a variety of changes have been made to


the classification of adenocarcinoma in hopes of
creating a more clinically relevant system. It should
be noted that as knowledge of molecular biology of
adenocarcinoma continues to grow and novel thera-
peutic agents are developed, it is likely that the
classification will evolve. There are additionally
several areas that need additional study, particularly
as the vast majority of information is gleaned from
studies of low stage tumors. Additionally, how the
IASLC classification will be incorporated into the
forthcoming revision of the WHO classification is Fig. 5 Squamous cell carcinoma with classic features of
keratin pearl formation in the center of a tumor nest (H and
currently in progress. Finally, the updated classifica- E 4009)
tion post-dates the 7th edition of the AJCC staging
guidelines and this will need to be rectified. In par-
ticular, the implications for staging in regard to ADC of tumors with so-called ‘‘non-squamous’’ his-
tumors with prominent lepidic growth which fall tology (Scagliotti et al. 2008, 2009a, b).
outside the category of MIA is unclear and further On conventional hematoxylin and eosin (H and E)
study is needed to determine if the stage should be stained sections, discriminating between SCC and
based on overall tumor size or the size of the invasive ADC is usually straightforward; however, ancillary
component alone. methods may be needed to discriminate between the
two when overt features of differentiation are lacking.
Several studies have evaluated various immunohis-
3 Squamous Cell Carcinoma tochemical techniques in regard to discriminating
SCC and ADC. While there is no consensus on which
SCC is now the second most common type of lung combination is most optimal, in general, a panel
carcinoma behind ADC, for reasons that are not consisting of p63, CK5/6, thyroid transcription factor-
entirely clear. From a pathologic standpoint, features 1 (TTF-1) and a mucin stain such as mucicarmine
definitively supporting squamous differentiation will resolve the majority of cases, although ulti-
include overt keratinization or intercellular bridge mately a small percentage will remain unclassifiable.
formation (Fig. 5). In small biopsies, these features A staining pattern of p63 positive, CK5/6 positive,
may not be obvious, particularly in poorly- TTF-1 negative and mucin negative supports a diag-
differentiated tumors, and it has become more critical nosis of SCC while the reverse staining pattern
to accurately separate SCC from ADC for optimal supports a diagnosis of ADC (Wu et al. 2003; Camilo
patient management. This is primarily due to treat- et al. 2006; Kargi et al. 2007; Loo et al. 2010;
ment management in regard to chemotherapy. Nicholson et al. 2010; Terry et al. 2010). A small
Patients with SCC should not receive the vascular percentage of ADC will show focal staining for p63
endothelial growth factor (VEGF) inhibitor bev- and, unless the finding is extensive, this should not be
acizumab due to the increased risk of life-threatening taken as evidence of co-existent squamous differen-
hemorrhage (Cohen et al. 2007). Conversely, EGFR tiation in the presence of diffuse TTF-1 positivity
tyrosine kinase inhibitors such as erlotinib and gefi- (Terry et al. 2010). Additionally, while mucinous
tinib are primarily recommended for patients with ADC is generally not easily confused with SCC, it
advanced ADC harboring EGFR mutations, particu- should be noted that mucinous ADC are generally
larly those in exons 19 or 21 (Mitsudomi et al. 2005; TTF-1 negative but these tumors will be positive for
Mok et al. 2009; Maemondo et al. 2010). Addition- mucin stains and negative for p63 and CK5/6 (Rossi
ally, pemetrexed is recommended for patients with et al. 2004; Travis et al. 2011).
Pathology of Lung Cancer 59

preparation in order to perform ancillary testing. Such


4 Molecular Testing in Lung Cancer testing cannot be consistently or reliably performed
on smear preparations in most institutions. As such,
The advent of targeted therapy has placed greater communication between the radiologist and patholo-
emphasis on molecular testing for the optimal treat- gist is of extreme importance in the setting of fine
ment of lung cancer, particular in patients with needle aspirations in particular to try to ensure that as
advanced disease. EGFR mutations are of particular much material as possible is submitted for a cell block
interest due to the response of tumors harboring in order to maximize the potential for ancillary testing
mutations, particularly those in exons 19 and 21, to (Rekhtman et al. 2011; Travis et al. 2011).
EGFR tyrosine kinase inhibitors such as gefitinib and In general, both tissue biopsies and cytology
erlotinib (Mitsudomi et al. 2005; Dacic 2008; Mok materials submitted for cell block are suitable for
et al. 2009; Maemondo et al. 2010; Mitsudomi et al. molecular testing. Specimens should ideally be fixed
2010). Conversely, tumors harboring K-RAS muta- in 10% neutral buffered formalin and acidic fixatives
tions are generally unresponsive to these agents or those containing heavy metals such as Bouin’s,
(Dacic 2008; Dacic et al. 2010). EML4-ALK trans- B-5 or Zenker’s are to be avoided as these inhibit
locations are also of particular interest as their pres- molecular analysis. Similarly, tissue which has been
ence is associated with response to crizotinib (Soda decalcified is inappropriate for molecular testing
et al. 2007). Other mutations which are not frequently (Gillespie et al. 2002; Srinivasan et al. 2002; Pirker
found in lung carcinomas but are of increasing clini- et al. 2010). All small biopsies pose similar issues of
cal interest include BRAF and PIK3CA HER2, and tissue management whether small tissue biopsies or
MET, among others. While there has been some cytology cell blocks, with a primary difference
correlation with clinical features and certain histo- being that tumor enrichment via microdissection can
logic types as discussed above, these features alone be performed more easily on tissue specimens as
cannot be used to select patients for mutation testing opposed to cell blocks. Otherwise, while sensitivity
(Mitsudomi et al. 2005; Dacic 2008; Mok et al. 2009; and specificity may vary with the precise sequencing
Douillard et al. 2010). technique used, a specimen should optimally contain
Testing for these genetic alterations generally a minimum of 500 tumor cells and tumor cells should
requires either DNA sequencing (EGFR, KRAS) or comprise at least 25% of the specimen, although some
fluorescence in situ hybridization (EML4-ALK). As authors have recommended 50% as an ideal minimum
such, in addition to adequate material for potential (Pirker et al. 2010). Recognition of these require-
ancillary studies for tumor subtyping as discussed ments and development of an appropriate strategy for
above, adequate material must be available for optimizing the amount of tumor obtained at the time
molecular testing as well in order to optimize patient of biopsy should maximize the amount of information
therapy (Dacic 2008; Pirker et al. 2010; Travis et al. that the pathologist is able to provide from a small
2011). As the majority of lung cancers are not sample and minimize the need to obtain addition
resected and are diagnosed by small biopsy or cytol- tissue for the sole purpose of molecular testing.
ogy specimens, it is important for the pathologist to In conclusion, there is increasing importance in
develop an appropriate strategy for optimizing tissue accurate subclassification of tumors formerly lumped
for ancillary and molecular testing. together as NSCLC, as well as an expanding need for
Biopsy specimens typically received from radiol- molecular testing. As most lung cancers are diagnosed
ogists include needle core biopsies as well as fine on small biopsies or cytology specimens, frequently
needle aspirations. While both types of specimens obtained from radiologists, the pathologist is required
yield small amounts of material and thus pose similar to ‘‘do more with less’’. Appropriate communication
issues in management for additional studies, com- between the pathologist and radiologist in regard to a
munication is particularly critical for fine needle strategy for optimal tissue management is essential
aspirations. For needle aspiration cytology specimens, for appropriate patient management (Travis et al.
adequate material must be present in a cell block 2011).
60 M. B. Beasley

(Fig. 6a, b). Typically a region of necrosis is sur-


rounded by a rim of foamy macrophages and inflam-
matory cells. Cholesterol cleft formation may be
present and a giant cell reaction may occur. Residual
tumor cells tend to have greater cytologic atypia and
pleomorphism in comparison to corresponding pre-
treatment specimens. The pattern of response present
does not appear to be related to the type of neo-adjuvant
therapy administered (i.e., chemotherapy plus radiation
therapy vs. chemotherapy alone), or with the type of
chemotherapy administered. Squamous carcinomas
tend to show a significantly greater degree of response
than adenocarcinomas (Junker et al. 1997a, b, 2001;
Liu-Jarin et al. 2003). Although the degree of fibrosis
tends to correlate with radiographic evidence of size
reduction, radiologic evidence of size regression does
not seem to significantly correlate with the degree of
histologic tumor response (Liu-Jarin et al. 2003).

References

Amin MB, Tamboli P et al (2002) Micropapillary component in


lung adenocarcinoma: a distinctive histologic feature with
possible prognostic significance. Am J Surg Pathol 26(3):
358–364
Beasley MB, Brambilla E et al (2005) The 2004 World Health
Fig. 6 Adenocarcinoma following pre-operative radiation and Organization classification of lung tumors. Semin Roentgenol
chemotherapy. A focus of residual adenocarcinoma is present 40(2):90–97
in the lower left, while the adjacent tissue shows hemorrhage, Borczuk AC (2009) Micropapillary histology: a frequent
fibrosis, inflammatory infiltrates and occasional foamy macro- morphology of mutation-associated lung adenocarcinoma?
phages (a). Elsewhere, viable tumor is absent and only foamy Am J Clin Pathol 131(5):615–617
macrophages and necrosis are present (b) (H and E 4009) Borczuk AC, Qian F et al (2009) Invasive size is an
independent predictor of survival in pulmonary adenocar-
cinoma. Am J Surg Pathol 33(3):462–469
Brownlee NA, Mott RT et al (2005) Mucinous (colloid)
adenocarcinoma of the lung. Arch Pathol Lab Med 129(1):
5 Pathology of Lung Carcinomas 121–122
Treated With Radiation and/ Camilo R, Capelozzi VL et al (2006) Expression of p63, keratin
or Neo-adjuvant Therapy 5/6, keratin 7, and surfactant-A in non-small cell lung
carcinomas. Hum Pathol 37(5):542–546
Cohen MH, Gootenberg J et al (2007) FDA drug approval
Administration of neo-adjuvant therapy prior to the
summary: bevacizumab (Avastin) plus Carboplatin and
resection of pulmonary carcinomas has become more Paclitaxel as first-line treatment of advanced/metastatic recur-
commonplace and is particularly aimed at stage IIIA rent nonsquamous non-small cell lung cancer. Oncologist
carcinomas. Initial studies suggest that tumors exhib- 12(6):713–718
Copin MC, Buisine MP et al (2001) Mucinous bronchioloal-
iting a complete histologic response or those that show
veolar carcinomas display a specific pattern of mucin gene
extensive response, with\10% of the gross tumor mass expression among primary lung adenocarcinomas. Hum
containing histologically viable tumor, have a survival Pathol 32(3):274–281
advantage (Junker et al. 1997a, b, 2001). Histologic Dacic S (2008) EGFR assays in lung cancer. Adv Anat Pathol
15(4):241–247
evidence of tumor response consists of coagulative or
Dacic S, Shuai Y et al (2010) Clinicopathological predictors of
infarct-like necrosis, fibrosis, foam cell and/or giant EGFR/KRAS mutational status in primary lung adenocar-
cell infiltration and mixed inflammatory cell infiltrates cinomas. Mod Pathol 23(2):159–168
Pathology of Lung Cancer 61

Douillard JY, Shepherd FA et al (2010) Molecular predictors of adenocarcinoma (\/=20 mm) with mixed bronchioloalveo-
outcome with gefitinib and docetaxel in previously treated lar and invasive subtypes (Noguchi’s type C tumours).
non-small-cell lung cancer: data from the randomized phase Histopathology 46(6):677–684
III INTEREST trial. J Clin Oncol 28(5):744–752 Mitsudomi T, Kosaka T et al (2005) Mutations of the epidermal
Finberg KE, Sequist LV et al (2007) Mucinous differentiation growth factor receptor gene predict prolonged survival after
correlates with absence of EGFR mutation and presence of gefitinib treatment in patients with non-small-cell lung
KRAS mutation in lung adenocarcinomas with bronchi- cancer with postoperative recurrence. J Clin Oncol 23(11):
oloalveolar features. J Mol Diagn 9(3):320–326 2513–2520
Gillespie JW, Best CJ et al (2002) Evaluation of non-formalin Mitsudomi T, Morita S et al (2010) Gefitinib versus cisplatin
tissue fixation for molecular profiling studies. Am J Pathol plus docetaxel in patients with non-small-cell lung cancer
160(2):449–457 harbouring mutations of the epidermal growth factor
Hata A, Katakami N et al (2010) Frequency of EGFR and receptor (WJTOG3405): an open label, randomised phase
KRAS mutations in Japanese patients with lung adenocar- 3 trial. Lancet Oncol 11(2):121–128
cinoma with features of the mucinous subtype of bronchi- Miyoshi T, Satoh Y et al (2003) Early-stage lung adenocarcinomas
oloalveolar carcinoma. J Thorac Oncol 5(8):1197–1200 with a micropapillary pattern, a distinct pathologic marker for a
Hoshi R, Tsuzuku M et al (2004) Micropapillary clusters in significantly poor prognosis. Am J Surg Pathol 27(1):101–109
early-stage lung adenocarcinomas: a distinct cytologic sign Mok TS, Wu YL et al (2009) Gefitinib or carboplatin-paclitaxel in
of significantly poor prognosis. Cancer 102(2):81–86 pulmonary adenocarcinoma. N Engl J Med 361(10):947–957
Inamura K, Satoh Y et al (2005) Pulmonary adenocarcinomas Nicholson AG, Gonzalez D et al (2010) Refining the diagnosis
with enteric differentiation: histologic and immunohisto- and EGFR status of non-small cell lung carcinoma in biopsy
chemical characteristics compared with metastatic colorec- and cytologic material, using a panel of mucin staining,
tal cancers and usual pulmonary adenocarcinomas. Am J TTF-1, cytokeratin 5/6, and P63, and EGFR mutation
Surg Pathol 29(5):660–665 analysis. J Thorac Oncol 5(4):436–441
Junker K, Thomas M et al (1997a) Tumour regression in non- Noguchi M, Morikawa A et al (1995) Small adenocarcinoma of
small-cell lung cancer following neoadjuvant therapy. the lung. Histologic characteristics and prognosis. Cancer
Histological assessment. J Cancer Res Clin Oncol 75(12):2844–2852
123(9):469–477 Pirker R, Herth FJ et al (2010) Consensus for EGFR mutation
Junker K, Thomas M et al (1997b) [Regression grading of testing in non-small cell lung cancer: results from a
neoadjuvant non-small-cell lung carcinoma treatment]. European workshop. J Thorac Oncol 5(10):1706–1713
Pathologe 18(2):131–140 Rekhtman N, Brandt SM et al (2011) Suitability of thoracic
Junker K, Langner K et al (2001) Grading of tumor regression cytology for new therapeutic paradigms in non-small cell
in non-small cell lung cancer : morphology and prognosis. lung carcinoma: high accuracy of tumor subtyping and
Chest 120(5):1584–1591 feasibility of EGFR and KRAS molecular testing. J Thorac
Kamiya K, Hayashi Y et al (2008) Histopathological features Oncol 6(3):451–458
and prognostic significance of the micropapillary pattern in Rodig SJ, Mino-Kenudson M et al (2009) Unique clinicopath-
lung adenocarcinoma. Mod Pathol 21(8):992–1001 ologic features characterize ALK-rearranged lung adeno-
Kargi A, Gurel D et al (2007) The diagnostic value of TTF-1, carcinoma in the western population. Clin Cancer Res
CK 5/6, and p63 immunostaining in classification of lung 15(16):5216–5223
carcinomas. Appl Immunohistochem Mol Morphol Rossi G, Murer B et al (2004) Primary mucinous (so-called
15(4):415–420 colloid) carcinomas of the lung: a clinicopathologic and
Kawakami T, Nabeshima K et al (2007) Micropapillary pattern and immunohistochemical study with special reference to
grade of stromal invasion in pT1 adenocarcinoma of the lung: CDX-2 homeobox gene and MUC2 expression. Am J Surg
usefulness as prognostic factors. Mod Pathol 20(5):514–521 Pathol 28(4):442–452
Kuroda N, Hamaguchi N et al (2006) Lung adenocarcinoma Sakurai H, Dobashi Y et al (2004a) Bronchioloalveolar
with a micropapillary pattern: a clinicopathological study of carcinoma of the lung 3 centimeters or less in diameter: a
25 cases. APMIS 114(5):381–385 prognostic assessment. Ann Thorac Surg 78(5):1728–1733
Lim E, Goldstraw P et al (2008) Proceedings of the IASLC Sakurai H, Maeshima A et al (2004b) Grade of stromal invasion in
International Workshop on Advances in Pulmonary Neuro- small adenocarcinoma of the lung: histopathological minimal
endocrine Tumors 2007. J Thorac Oncol 3(10):1194–1201 invasion and prognosis. Am J Surg Pathol 28(2):198–206
Liu-Jarin X, Stoopler MB et al (2003) Histologic assessment of Sartori G, Cavazza A et al (2009) EGFR and K-ras mutations
non-small cell lung carcinoma after neoadjuvant therapy. along the spectrum of pulmonary epithelial tumors of the
Mod Pathol 16(11):1102–1108 lung and elaboration of a combined clinicopathologic and
Loo PS, Thomas SC et al (2010) Subtyping of undifferentiated molecular scoring system to predict clinical responsiveness
non-small cell carcinomas in bronchial biopsy specimens. to EGFR inhibitors. Am J Clin Pathol 131(4):478–489
J Thorac Oncol 5(4):442–447 Scagliotti GV, Parikh P et al (2008) Phase III study comparing
Maemondo M, Inoue A et al (2010) Gefitinib or chemotherapy cisplatin plus gemcitabine with cisplatin plus pemetrexed in
for non-small-cell lung cancer with mutated EGFR. N Engl chemotherapy-naive patients with advanced-stage non-
J Med 362(25):2380–2388 small-cell lung cancer. J Clin Oncol 26(21):3543–3551
Makimoto Y, Nabeshima K et al (2005) Micropapillary pattern: Scagliotti G, Hanna N et al (2009a) The differential efficacy of
a distinct pathological marker to subclassify tumours with a pemetrexed according to NSCLC histology: a review of two
significantly poor prognosis within small peripheral lung Phase III studies. Oncologist 14(3):253–263
62 M. B. Beasley

Scagliotti GV, Park K et al (2009b) Survival without toxicity squamous cell carcinomas in small tumor samples. Am J
for cisplatin plus pemetrexed versus cisplatin plus gemcit- Surg Pathol 34(12):1805–1811
abine in chemonaive patients with advanced non-small cell Travis WD, Brambilla E et al (2004) Pathology and genetics:
lung cancer: a risk-benefit analysis of a large phase III tumours of the lung, pleura, thymus and heart. IARC, Lyon
study. Eur J Cancer 45(13):2298–2303 Travis WD, Brambilla E et al (2011) International association
Soda M, Choi YL et al (2007) Identification of the transforming for the study of lung cancer/american thoracic society/
EML4-ALK fusion gene in non-small-cell lung cancer. european respiratory society international multidisciplinary
Nature 448(7153):561–566 classification of lung adenocarcinoma. J Thorac Oncol
Srinivasan M, Sedmak D et al (2002) Effect of fixatives and 6(2):244–285
tissue processing on the content and integrity of nucleic Wu M, Wang B et al (2003) p63 and TTF-1 immunostaining. A
acids. Am J Pathol 161(6):1961–1971 useful marker panel for distinguishing small cell carcinoma
Suzuki K, Asamura H et al (2002) ‘‘Early’’ peripheral lung of lung from poorly differentiated squamous cell carcinoma
cancer: prognostic significance of ground glass opacity on of lung. Am J Clin Pathol 119(5):696–702
thin-section computed tomographic scan. Ann Thorac Surg Yim J, Zhu LC et al (2007) Histologic features are important
74(5):1635–1639 prognostic indicators in early stages lung adenocarcinomas.
Terry J, Leung S et al (2010) Optimal immunohistochemical Mod Pathol 20(2):233–241
markers for distinguishing lung adenocarcinomas from
Radiologic Imaging of Lung Cancer
Palmi Shah and James L. Mulshine

Contents Abstract
Recent advances in technology like faster, high
1 Introduction.............................................................. 63 resolution CT scanners, new data in lung cancer
2 Overview ................................................................... 64 screening combined with many changes in the
classification, staging and novel therapies for lung
3 Radiologic Imaging.................................................. 64
3.1 Detection .................................................................... 64 cancer are redefining the role imaging plays in
3.2 Staging ....................................................................... 68 detection, staging and management of the disease.
3.3 PostTherapy Imaging................................................. 72 This chapter describes the utility and limitations of
4 Conclusions ............................................................... 72 the different radiological modalities in various
stages of the disease. Since it is the mainstay in
5 Future........................................................................ 72
lung cancer management emphasis is placed on the
References.......................................................................... 73 application of CT imaging. PET-CT has been
discussed separately in this volume.

1 Introduction

Lung cancer rapidly rose in incidence in the 20th


century and became the leading cause of cancer-related
mortality. As we move into the 21st century the inci-
dence is unlikely to decline secondary to the rising
number of cases in the developing world (Toh 2009).
Challenges associated with identifying, diagnosing and
treating the disease are reflected by the steady 5-year
mortality of 13% in the last 15 years.
Lung cancer was the most commonly diagnosed
cancer as well as the leading cause of cancer death in
males in 2008 globally. Among females, it was the fourth
P. Shah most commonly diagnosed cancer and the second lead-
Department of Radiology, Rush University,
ing cause of cancer death globally. It accounted for 13%
Chicago, IL, USA
(1.6 million) of the total cases and 18% (1.4 million) of
J. L. Mulshine (&)
the deaths in 2008 globally (Jemal et al. 2011).
Department of Internal Medicine,
Rush University, Chicago, IL, USA Lung cancer remains the leading cause of cancer-
e-mail: James_L_Mulshine@rush.edu related death in the United States.

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 63
DOI: 10.1007/174_2011_267, Ó Springer-Verlag Berlin Heidelberg 2011
64 P. Shah and J. L. Mulshine

Rapid technological advances in the 21st century these cancers and to introduce more immunohisto-
have contributed to the now validated value of chemical staining to keep up with the new novel
imaging in lung cancer from screening, diagnosis, targeted therapies now available.
staging, therapy planning to treatment monitoring. Clinical presentations of these various subtypes of
Advancements in multi-detector CT (Computed lung cancer are varied and overlapping. In addition it is
Tomography), MRI (Magnetic Resonance Imaging), believed that 25% of patients with lung cancer are
PET (Positron Emission Tomography), now inte- asymptomatic at the time of diagnosis while the
grated PET- CT in conjunction with endobronchial symptomatic patients present with symptoms ranging
and transesophageal ultrasound have further enhanced from mild cough, hemoptysis or chest pain to seizures,
and refined the vital role imaging plays in disease dependant on tumor location and tumor spread.
detection and management. Correspondingly, the imaging findings of these
The staging and management of the disease is a tumors similarly are wide and varied and knowledge
multidisciplinary process with multiple clinical and of these would facilitate in timely diagnosis, appro-
nonclinical modalities playing a complimentary role priate imaging and help guide optimal management
with each other. The utility of each modality is getting strategies.
better defined with the push for earlier detection, need As a background to the discussion of imaging it is
for accurate staging and the need to complement the important to introduce the new TNM classification of
newer and varied therapy choices available today. lung cancer (7th edition) released by the American
In this chapter we will discuss the role of radiologic joint committee on cancer in 2010 (AJCC 2010) along
imaging in lung cancer with emphasis on CT. PET-CT with the revised nomenclature for lymph nodes as
which also constitutes a mainstay of lung cancer detailed in the international association for the study
management is used for initial radiologic staging and of lung cancer (IASLC) lymph node map. The new
also plays a significant role in the evaluation of tumor staging classification elucidates stage clustering that
recurrence and treatment monitoring, is discussed in a is more homogenous in their clinical behavior and
separate chapter and will not be detailed here. outcome.

2 Overview 3 Radiologic Imaging

The 2004 World health organization (WHO) (Travis Various modalities with their imaging manifestations
et al. 2004) classification of primary lung tumors have been listed under the following outline, which
divided them by their light microscopy features into- follows the course of the disease.
small-cell cancer and non-small-cell cancer Detection
(NSCLC). NSCLC includes adenocarcinoma includ- Staging
ing bronchioloalveolar cell carcinoma (BAC), squa- Recurrence
mous cell carcinoma and large cell carcinoma.
A new pathological classification for adenocarci-
nomas has just been introduced in which the terms 3.1 Detection
BAC and mixed subtype adenocarcinoma are no
longer used. For resection specimens, new concepts 3.1.1 Chest Radiographs
are introduced such as adenocarcinoma in situ (AIS) These form the frontline and the most frequently used
and minimally invasive adenocarcinoma (MIA). AIS modality for evaluation of symptoms related to the
and MIA are usually nonmucinous but rarely may be cardiopulmonary system.
mucinous (Travis et al. 2011). The term BAC has still Frequently lung cancers are incidentally diagnosed
been used here for description because of familiarity, on chest radiographs done for an unrelated reason.
however it’s use in the future is discouraged. Nodules and masses may be detected. It is rare to
Invasive adenocarcinomas are subclassified by the detect a nodule less than 1 cm on chest radiographs
predominant pattern after using comprehensive his- secondary to limitations in sensitivity (Kundel 1981).
tologic subtyping to recognize the heterogeneity of When detected the first step involves the search for
Radiologic Imaging of Lung Cancer 65

older studies. If upon review with old studies it is screening studies. The Mayo Chest Radiography
established that the nodule has been stable in size for Lung Cancer Screening Project (Muhm et al. 1983)
2 years, it is likely benign. showed that 45 of the 50 peripheral lung cancers were
Nodules or masses may be peripheral or central. initially missed, and 12 of the 16 central cancers were
Peripheral nodules and masses are easier to detect due visualized in retrospect but were overlooked initially.
to the more favorable signal to noise ratio when the These results reflect the challenge of visualizing early
lesion is silhouetted by the air filled normal lung tis- lung cancer given the limited capabilities of chest
sue. In this location detected cancers are more com- x-ray detection. Another study by Heelan et al. found
monly adenocarcinomas. that 65% of cancers were missed on the initial chest
On radiographs the nodule may display a range of radiograph (Heelan et al. 1989). More recently the
features including ill defined/spiculated margins or a preliminary result from the NLST screening trial
thick-walled cavity. Squamous cancers more fre- also found a statistically significant difference in the
quently are found centrally and often present as cav- sensitivity of CT scans to detect lung cancers in
itating lesions. Eccentric or stippled calcification may comparison to chest radiographs (National Cancer
be present, and this presentation is more common Institute 2010).
with adenocarcinomas. All other forms of calcifica- In contrast to the disappointing chest-x-ray
tion are considered benign. screening studies, a recent report has suggested that
Bronchioloalveolar cell carcinoma—mucinous the true lung cancer mortality reduction from spiral
form can present with segmental or lobar opacity based screening to be over 30% and possibly as high
like pneumonia. They can also present as single or as 60% based on computer modeling or other com-
multiple pulmonary nodules. parative approaches (Henschke et al. 2010). Given the
Central lesions can present as hilar masses, which legacy of lung cancer screening, research into defin-
adds to the opacity at the hilum. These cancers may ing the optimal approach to deliver effective lung
result in compromise of airflow and therefore are cancer screening services is essential in allowing cost
often associated with non-resolving pneumonias, effective and robust lung cancer screening, to be
consolidations or persistent atelectasis. These pro- implemented and allow sustained lung cancer mor-
cesses are often in a lobar or segmental distribution. tality reduction.
In addition the functional consequence of a large
central lesion is the finding of bronchial cut off on the 3.1.2 CT Scans
radiographs. This results in either complete atelectasis or As with chest radiographs when evaluating for early
partial obstruction. These changes may result in a lung cancer many new cancers are now found to be
hyperlucent lung ipsilaterally. Central lesions are more located in the periphery of the lung. It has been pro-
common with small-cell and squamous cell carcinomas. posed that this location is the result of small particle
In regionally advanced cancer associated with size of tobacco consumption products that are being
mediastinal invasion and mediastinal lymph node emitted from the end of filtered cigarettes. However
involvement a variety of findings including a widened this air-filled space is a particularly favorable location
mediastinum, thickened right paratracheal stripe, loss for x-ray-based imaging.
of concavity of the aortopulmonary window, con- The widespread availability, faster speed, better
vexity of the mediastinum and splaying of the carina contrast, increased specificity and the exquisite sen-
have been described. sitivity of CT scanners make them the primary
More advanced disease can present with rib/osse- modality for the initial evaluation of solitary pul-
ous destruction in association with peripheral tumors, monary nodules, mediastinal or hilar masses.
pleural effusions or thickening. Linear opacities Sometimes lung cancers are detected on the inci-
radiating from the mass to the periphery or diffuse dental lung images of CT scans of contiguous body
interstitial thickening can be seen with lymphangitic parts or CT done to evaluate other non-specific
carcinomatosis. Metastatic osseous involvement may abnormalities seen initially on radiographs.
also be visible on the radiograph. Low-dose CT screening has attracted considerable
Multiple studies have shown a poor detection rate recent attention in the wake of the recent release of
for early lung cancer on chest radiographs even in the preliminary findings of the National Lung Cancer
66 P. Shah and J. L. Mulshine

Screening trial (NLST). The NCI released to the press differentiation between benign and malignant nod-
that their researchers had found, in interim analyses, ules. Nodules with volume doubling time of less than
20.3% fewer lung cancer deaths among those who 400 days were viewed as suspicious and further
were screened with low-dose helical CT compared evaluated for malignancy. This study stated detection
with those who were screened with chest x-rays. sensitivity of 95% and a specificity of 99% for
This was the magnitude of benefit that was the nodules with volumes between 50 and 500 mm3
basis of the trial design of the NSLT since it was (van Klaveren et al. 2009).
thought that reducing lung cancer mortality by 20% Margin evaluation also helps in increasing the spec-
was consistent with the threshold for a significant ificity for detection of lung cancer. A lobulated margin
screening benefit. indicates uneven growth and is a finding that increases
It is important to emphasize that the data derived the potential for lung cancer (Libby et al. 2004).
from the NLST was obtained from a very specific Spiculated margins in adenocarcinomas are known
population group—individuals at high risk for to confer a worse prognosis in comparison to lobu-
developing lung cancer due to present or past heavy lated margins (Aoki et al. 2001).
smoking, aged 55–74, and the observed mortality The presence of ‘‘corona radiata’’, multiple strands
benefit may not necessarily apply to the general extending from the tumor, indicating tumor extension
population (The National Lung Screening Trial 2010). or fibrotic response is best seen on CT (Klein and
As we await more definitive release of the full results Braff 2008a, b). Pleural tail and tethering are addi-
from the trial many are hopeful that we are looking at tional features highly concerning for tumor.
a shift in paradigm in the struggle to detect the cancer Calcification may be present in 6–10% of
early and subsequent decrease in lung cancer-related bronchogenic carcinomas on CT (Zerhouni et al.
mortality. 1986). Both small and non-small-cell carcinomas can
show calcification, which is typically amorphous,
3.1.2.1 Peripheral Tumors stippled or cloud like. Eccentric calcifications are also
Solitary pulmonary solid nodules and imaging features identified in lung cancers and usually represent
of lung cancer: granulomas engulfed by the carcinoma.
Characterization of solitary pulmonary nodules is Other features like convergence of vessels, inva-
enhanced by CT scans. Non-contrast CT is usually sion of pulmonary vasculature, internal necrosis,
adequate. Some reports have suggested that the use of cavitation (usually squamous cell cancers) with thick
contrast may enhance nodule detection and can help shaggy walls ([15 mm) all contribute to increasing
in increasing specificity for lung cancers. This suspicion for malignancy and are more reliably
approach incurs considerable increase in cost, the assessed on CT scans.
potential of renal failure related to the osmotic load of
the contrast agent. Also due to recent advances the Groundglass density/nonsolid/part solid nodules:
proposed benefit of this approach has been superseded Screening CT data has revealed that a high number of
by PET-CT. malignant nodules can have ground glass density
Lung cancer nodule detection is complicated by (Henschke et al. 2002). These tend to be adenocar-
the occurrence of a large number of pulmonary nod- cinomas with bronchioloalveolar cell component or
ules that can be frequently found in the lung of bronchioloalveolar carcinomas.
smokers. An approach to permit the differentiation of These nodules may be purely groundglass in den-
nodules that are benign from those having malignant sity (nonsolid nodules) or may have central solid
potential is to measure the growth of this nodules component, when they are termed, part solid nodules.
across a time interval. Studies have shown that for nodules less than 3 cm, if
The classical criteria for malignancy in a solitary the groundglass component is larger than 50%, there
pulmonary nodule at baseline CT is size greater than was a lower incidence of vessel invasion and regional
3 cm (90%). A recent study used increase in volume lymph node spread (Aoki et al. 2001). These patients
of 25% or more measured using non proprietary also had a statistically significant better prognosis
software extrapolated to calculate estimated volume than patients with nodules with a greater than 50%
doubling time, as a measure to allow for greater solid component (Aoki et al. 2000).
Radiologic Imaging of Lung Cancer 67

using the CAD system (P \ 0.1), with a true positive


rate of 94% (Awai et al. 2004).
More importance is expected in the future although
as of now visual assessment and identification of
growth by cross-sectional diameter remains the
mainstay for assessment for change (Padhani and
Ollivier 2001).
3.1.2.2 Central Tumors
Most unilateral mediastinal masses are lung cancers
in adults. Use of IV (intravenous) iodinated contrast is
Fig. 1 a,b,c Three different patients showing a Spiculated preferred in the evaluation of central lesions, for
nodule b Lobulated nodule c Groundglass/non solid nodule
detecting vascular invasion and evaluating mediasti-
nal involvement.
Of particular note is that these tumors may show Small and squamous cell carcinomas occur more
no or low metabolic activity on PET imaging and frequently in the central location and because of their
should be viewed with suspicion in spite of the PET proximity to the central airways and vessels can cause
appearance. postobstructive atelectasis and consolidations/pneu-
Resection is usually recommended in a high-risk monias associated with hilar enlargement.
patient (Klein and Braff 2008a, b). Contrast-enhanced CT scan (CECT) may be useful
Air bronchograms and cystic/bubbly lucencies in evaluation of these tumors as the contrast helps in
within the nodule are more frequently associated with delineating the tumor from surrounding atelectatic
adenocarcinomas with bronchioloalveolar cell com- changes. The tumor usually enhances less than the
ponent and bronchioloalveolar carcinomas (Kuriyama surrounding atelectasis. This is helpful in planning
et al. 1991; Zwirewich et al. 1991 (Fig. 1). radiotherapy and estimating tumor volume. The ben-
Rate of growth of nodules is important to assess for efits of using contrast in thoracic imaging should be
establishment of stability, which would indicate a evaluated relative to the issues of increased cost and
benign process. The absence of growth of a nodule the potential of iodine-related side effects as well as
over a period of 2 years is generally an indicator of a the potential for renal complications especially in the
benign process. Calculated as nodule doubling time setting of an elderly patient with underlying renal
we do know that benign processes have doubling time compromise.
of less than 30 days or more than 450 days (Gorlova Additional findings of alteration of shape of lobar
et al. 2005). consolidation or atelectasis secondary to tumor bulk,
Although once considered reliable we now know like bulging of fissures may indicate underlying
that some of the ground glass nodules can grow at a neoplasm.
much slower rate and these require long-term follow- Findings of airway narrowing, displacement or
up (Yankelevitz et al. 1997). obstruction would also suggest an underlying mass.
Many computer aided detection (CAD) systems A central mass may be visible. Persistent consolida-
are available for improving detection of nodules on tion for greater 2 weeks may also be suggestive of the
chest radiographs and CT scans, which serve as sec- presence of neoplastic disease (Fig. 2).
ond readers and help in enhancing the detection Endobronchial tumors may present as nondepen-
accuracy by the radiologist, who still remains the dant masses in the airway when small, or may be
primary reader. Newer systems for segmentation and associated with distal mucus plugging or segmental/
volumetric analysis of nodule size, increase accuracy lobar atelectasis when occlusive.
and reliability for assessing subtle nodule/mass Finally CT has a growing role in guiding needle
dimension changes to better evaluate response to and core biopsies from suspicious lesions, for tissue
therapy (Revel et al. 2004). Awat et al. demonstrated sampling, to establish the diagnosis of cancer or in
a statistically significant improvement in performance allowing research studies with regard to selecting
for lung nodule detection of all study participants personalized molecular therapies.
68 P. Shah and J. L. Mulshine

been established to reflect management options and


survival.
Refinements in the TNM classification of lung
cancers were released in the 7th edition of American
joint committee on cancer in 2010 based on a com-
prehensive reevaluation effort and were revised to
more accurately reflect survival in the different dis-
ease stages.
Lung carcinomas are still divided into small and
non-small-cell carcinomas for the marked differences
in natural history and response to therapy. Note
should be made that non-small-cell cancers, small-
cell cancers and carcinoids now have the same TNM
staging systems (AJCC 2010).
For the sake of radiotherapy planning small-cell
Fig. 2 CECT showing left upper lobe central tumor as carcinomas are classified into limited and extensive
hypodense area as compared to surrounding consolidation disease. Limited disease is confined to the same
hemithorax but also includes involvement of the
3.1.3 PET-CT contralateral mediastinal and supraclavicular lymph
The utility of evaluation of solitary pulmonary nodules nodes (Simon and Turrisi 2007). Imaging plays an
by PET-CT is well-established secondary to its high extended role in identifying extensive disease.
negative predictive value for lesions that are benign,
making it invaluable. This has been detailed elsewhere. 3.2.1 Chest Radiographs
The relative value of PET-CT compared to nodule There is a limited role these play in staging lung
growth rate determined by volumetric CT is an impor- cancers due to their poor sensitivity in detecting
tant area for further comparative effectiveness research. advanced disease. Mediastinal invasion is poorly
detected but may be suggested by an elevated dia-
3.1.4 MRI phragm, which would indicate phrenic nerve
Long scanning times, respiratory and cardiac motion involvement. An advanced stage may be suggested by
artifact and limited sensitivity to detect calcifications the size of the tumor by osseous destruction or
and endobronchial tumors renders MRI less useful in mediastinal widening on the initial evaluation.
evaluation for and of lung cancer as compared to CT.
It may have a role in evaluation of patients with 3.2.2 CT Scans
central tumors with surrounding atelectasis and con- The newer and faster spiral contrast-enhanced multi-
solidation in whom iodinated contrast is contraindi- slice computed tomography (CECT) offers exquisite
cated. The tumor shows different signal intensities on anatomic detail, and is the ideal choice to assess size
T2 weighted images and postgadolinium-enhanced of the tumor and its relationship to the surrounding
T1 weighted images as compared to the surrounding anatomical structures like the fissures and to the
parenchymal changes (Erasmus and Sabloff 2008). pleura and chest wall. Issues around quality control
MRI however plays a critical role in evaluation and measures to improve lung quantitation have been
and staging of Pancoast tumors secondary to its extensively discussed with the work of the Quantita-
ability to determine vertebral body, spinal and bra- tive Imaging Biomarker Alliance.
chial plexus invasion better. CT scans also play a big role in disease quantifi-
cation and helps in the initial radiological staging of
3.2 Staging the tumor. Usually they are performed prior to bron-
choscopy as detection of extensive disease may pre-
As with most cancers survival and prognosis in lung clude the need for the same. They are also helpful in
cancer depends on the cell type of the cancer and the determining therapy options and help in treatment
stage at which it is detected. The staging system has planning.
Radiologic Imaging of Lung Cancer 69

Accurate staging of the tumor is essential as ther- CECT or MRI may be necessary to accurately eval-
apeutic options depends on stage of the disease, in uate this as the primary lesion will enhance less than
addition to the cell type and the clinical and func- the surrounding atelectasis and the collapsed lung
tional status of the patient. may show hypodense mucus filled bronchi (UyBico
In the appropriate clinical context surgically et al. 2010).
resectable tumor is usually stage III A or lower for Endobronchial lesions more than 2 cm distal to the
non-small-cell cancers. Adjuvant radiation and che- carina also belongs in this category. These findings
motherapy are offered to certain stage IIA and III A are easily assessed on CT with the help of mutiplanar
disease and surgical options may be offered in IIIB reformatting.
disease if preoperative therapy downstages the tumor. Detection of a satellite nodule in the same lobe as
All non-surgical candidates are offered various com- the primary tumor and size greater than 7 cm would
binations of radiotherapy and chemotherapy depend- now shift the tumor to a T3 category. Endobronchial
ing on the nature of the cancer presentation. lesions less than 2 cm distal to the carina but not
Small-cell cancer comprises about 15% of lung involving the carina; tumors with local invasion of the
cancer, and is a neuroendocrine cancer, that is only chest wall, diaphragm, mediastinal pleura and parietal
treated surgically in the uncommon situation when it pericardium; superior sulcus tumors; and tumors with
presents as an isolated nodule. Chemotherapy with atelectasis and obstructive pneumonitis affecting the
radiotherapy is the mainstay of regionally confined entire lung are still considered stage T3 neoplasms
presentations but disseminated disease is generally (UyBico et al. 2010).
treated initially with chemotherapy only. Chest wall invasion may be suggested by rib ero-
Chest CT for suspected lung carcinoma should be sions, vertebral destruction and focal chest wall mass.
extended inferiorly to include the adrenal glands Detection of this alters the surgical technique. Some
secondary to the propensity of adrenal metastatic of the other CT signs described to suggest parietal
disease. The debate between a contrast versus non- pleura and chest wall invasion are obtuse angle
contrast study continues. For visualization of enlarged between tumor and pleura, loss of the extrapleural fat
hilar lymph node and 2R (high paratracheal) lymph plane, pleural thickening and extrapleural soft tissue.
node, a contrast study is more sensitive (Patz et al. Localised chest pain however still is the most specific
1999). A single study found no differences in detec- indicator of chest wall invasion (Akata et al. 2008).
tion of liver or adrenal metastases with or without CT and MRI are approximately equally sensitive and
contrast (Cascade et al. 1998). specific for the same varying from 63 to 90% and 83
Key factors in evaluation for staging would include to 86%.
staging the primary tumor (T category), intrathoracic The distinction between T3 and T4 lesions is
lymph node (N category) detection, distant pulmonary crucial as this draws the line between surgical versus
spread and extrathoracic spread (M category). non-surgical therapy (Fig. 3).
Primary tumor size and local and endobronchial Separate tumor nodules in the same lung but not in
spread determines the T category and primary tumor the same lobe as the primary lesion, which were
size is highly co-related with eventual clinical previously considered metastatic (M1) but due to
outcome. more favorable clinical behavior are now classified as
There are new subdivisions in the T1 and T2 T4 disease. T4 tumors may also invade the heart,
stages, which are determined by size. Tumor less than great vessels, trachea, carina, esophagus or vertebral
3 cm is T1 lesions while those between 3 and 7 cm body (UyBico et al. 2010).
are now categorized under the T2 category (UyBico Both CT and MRI, with multi-detector CECT
et al. 2010). being superior in most instances, detect mediastinal
Additional detection of visceral pleura invasion invasion. Clear encasement of vital structures like
may be suggested by puckering although more accu- trachea, esophagus, great vessels, etc. is evidence of
rate determination is made by pathology. Central T4 disease and mediastinal invasion.
tumors with associated atelectasis and obstructive Mere contact is not enough to suggest invasion and
pneumonitis extending to the hilar region but not circumferential contact of less than 90 degrees or
involving the entire lung are considered T2 tumors. preservation of mediastinal fat planes\3 cm or tumor
70 P. Shah and J. L. Mulshine

divisions provided by The IASLC Lung Cancer


Staging Project (Rusch et al. 2009).
Enlargement of a lymph node is the most validated
sign for metastatic involvement, however is not very
specific. A short axis dimension of 1 cm or greater is
a significant size and suspicious for metastatic dis-
ease, although the predictive accuracy of this criterion
is limited (Lau and Harpole 2000).
Additional changes including internal necrosis and
convexity of the lymph node and prescence of fatty
hilum are useful when present.
When using these criteria to determine lymph node
involvement the reported sensitivity and specificity of
CT is only about 50–65% (Arita et al. 1996; Primack
et al. 1994). The most accurate method of lymph node
Fig. 3 CECT showing bilateral enlarged mediastinal lymph staging still remains via surgery and mediastinal
nodes indicating at least N3 disease in NSLC. Subtle bony lymph node dissection. However the importance in
metastases in the vertebral body and small bilateral pleural
effusions
detection of these lymph nodes via CT is vital to help
guide sites of lymph node biopsy.
It is known that normal sized FDG negative
and pleura contact \3 cm are reliable indicators of mediastinal lymph nodes on CT and PET can have
non-invasion, but one should always be careful to malignant involvement in their mediastinal nodes.
exclude a patient from surgery based on CT criteria The ACCP guidelines state that invasive preoperative
alone (Bittner and Felix 1998; Hierholzer et al. 2000). mediastinal staging should be performed in these
In addition, the presence of a malignant pleural patients especially in the presence of central tumors
effusion, pleural dissemination or pericardial disease (Detterbeck et al. 2007).
now constitutes metastatic disease (M1a) and is no M category of the disease, especially extrathoracic
longer in the T category (UyBico et al. 2010). These disease may be assessed by dedicated CT of the abdo-
may be seen as complex pleural and pericardial men or contiguous CT to include the entire liver. These
effusions measuring more than fluid density on CT or are usually adequate to evaluate the liver and adrenal
may demonstrate the presence of internal-enhancing glands, constituting extrathoracic spread. One study
nodules or plaques. demonstrated no significant increase in detection of
The presence of localized interlobular thickening liver or adrenal metastases with contrast as compared to
which may be smooth or nodular with thickening of the a non-contrast examination (Cascade et al. 1998),
peribronchovascular interstitium usually starting however the debate to use or not to use routine iodin-
around the tumor could indicate lymphangitic carci- ated contrast for liver and adrenal metastases continues.
nomatosis, easily seen on CT scans even in early stages. Imaging the upper abdomen should be done as a
Contralateral pulmonary nodules also constitute routine in small-cell cancer secondary to the high
M1a disease and are detected on the lung windows of incidence of metastases at the time of initial diagnosis
the regular CT scan (UyBico et al. 2010). (Ravenel et al. 2010).
Intrathoracic lymph node staging determines the Adrenal nodules greater than 3 cm in size over-
nodal category (N) of the disease and is an important whelmingly are metastatic nodules. If the adrenal
predictor of outcome. Surgery is not considered in N3 nodule is indeterminate on the staging CT a dedicated
disease, which is constituted by contralateral lymph adrenal protocol contrast-enhanced CT or chemical
node enlargement and all significantly large lymph shift MRI can help further characterize this.
nodes in the ipsilateral or contralateral supraclavicular The same is true for liver metastases. Dedicated
or scalene stations. triple phase CT of the liver or postcontrast liver MRI
The position of the mediastinal and hilar lymph can be used as adjunct imaging in confounding cases
nodes is described according to the new map and (Ravenel et al. 2010).
Radiologic Imaging of Lung Cancer 71

3.2.4 MRI
The utility of MRI is limited in the evaluation of the
primary tumor except for imaging in Pancoast or
apical tumors. Here mutiplanar MRI with contrast
would be the imaging modality of choice given the
increased sensitivity in detecting spread to the bra-
chial plexus especially above T1, subclavian vessel
involvement and greater than 50% vertebral body
involvement all of which may make the tumor
inoperable.
Central tumors, which cannot be evaluated by CT
with iodinated contrast, could potentially be imaged
with MRI.
Chest wall invasion is equally well assessed by CT
Fig. 4 CECT showing left apical pancoast tumor with chest and MRI as detailed above.
wall invasion, rib and vertebral body destruction
Extrathoracic spread to the brain and spine are
better assessed with postcontrast MRI as compared to
Although postcontrast head CT can detect intra- CT. Sensitivity of MRI is significantly higher here.
cranial metastases, MRI with contrast remains the Routine staging MRI of the brain with contrast should
most sensitive examination. be considered in small-cell cancers secondary to the
Finally CT imaging is also used to guide in high frequency of intracranial metastases (Ravenel
radiofrequency ablation of tumors in patients who are et al. 2010).
poor surgical candidates (Fig. 4). Chemical shift imaging for adrenal nodules or
masses can be considered when CT is indeterminate.
3.2.3 Endoscopic Ultrasound Liver MRI could be utilized as a problem-solving tool
Newer techniques like endoscopic ultrasound can for indeterminate or discordant CT and PET-CT
evaluate lymph nodes in the vicinity of the esophagus, findings (Ravenel et al. 2010).
trachea or main bronchi, and therefore improve the
accuracy of endoscopic mediastinal lymph node 3.2.5 Bone Scans
sampling techniques. Endobronchial ultrasonography The utilization of bone scans for staging is progres-
(EBUS) can be performed to visualize and sample sively declining with the increased availability of
lymph node stations 2R/2L, 4R/4L and 7, as well as PET-CT, which has higher sensitivity and specificity
hilar stations. In short lymph node stations close to in evaluation of osseous metastases (Marom et al.
the trachea and the main bronchi can be sampled via 1999). If a PET-CT is not available or is not being
this technique. The lymph node stations sampled are considered a bone scan should be performed as part of
the same as those sampled by cervical mediastinos- the initial staging work up, especially in small-cell
copy (Vansteenkiste et al. 2010). cancer.
Esophageal ultrasonography (EUS) helps in visu- New onset bony pain, hypercalcemia, raised alka-
alization and sampling of the lymph node stations 4L, line phosphatase or pathologic fractures are usually
and levels 7, 8 and 9. This is complementary to the assessed by a bone scan during the course of the
other techniques, like endobronchial ultrasound and disease.
mediastinoscopy as lymph nodes at levels 8 and 9 Radiographs, CT scan or MRI of the affected
are not accessible by EBUS or mediastinoscopy region usually follows a positive bone scan.
(Vansteenkiste et al. 2010).
Published meta-analyses on EUS and EBUS gui- 3.2.6 V/Q (Ventilation and Perfusion) Scans
ded tissue diagnosis, reported a pooled sensitivity of Preoperative V/Q scan help quantify individual lung
90% and 94%, respectively, for CT-enlarged or PET- function to help in determining if the patient is a
positive mediastinal lymph nodes with a prevalence surgical candidate, and also in planning extent and
of malignant N2/3 disease of 68% (Gu et al. 2009). type of surgery to ensure that the patient can survive
72 P. Shah and J. L. Mulshine

with the residual postoperative lung function. Venti- is generally routine but the frequency of such studies
lation and perfusion fractions between the two lungs is not rigorously validated.
are estimated. CT is also used to detect second primary lesions in
these patients. Second metachronous primary tumor
3.2.7 PET-CT risk is estimated to occur in 1–2% patients per year
This youngest modality in the available technology (Johnson 1998).
for lung cancer imaging has emerged as the front-
runner for more accurate radiologic staging in bulky
primary lung cancer. This imaging tool can be fre- 4 Conclusions
quently used as one stop shopping for complete
evaluation of the tumor and the extent of disease. A CT scan is the time-tested modality playing a key role
statistically significant percentage of patients maybe in all aspects of the disease from screening, detection
upstaged or downstaged by the PET-CT scan. to staging and treatment monitoring. Multiple other
modalities and disciplines play complementary roles
in accurate preoperative and pretherapy staging of the
3.3 PostTherapy Imaging tumor helping to formulate the best management plan
for the patient.
Postradiation therapy imaging with CECT is subop- It should be emphasized that no patient should be
timal to evaluate response to therapy in most cases denied definitive treatment based on indeterminate
secondary to the surrounding changes which can imaging findings and when in doubt the answer would
simulate both radiation-induced fibrosis or spread of be to obtain histologic confirmation.
tumor. Some of the secondary signs, which may
indicate tumor response, are the actual diminished
size of tumor or internal necrosis posttreatment. 5 Future
Tumor recurrence within radiation-induced fibrosis is
particularly difficult to detect by CT in the early Low-dose chest CT screening for lung cancer in the
stages and PET-CT maybe a more sensitive exami- aim of reducing mortality may soon become an
nation for the same. accepted form of screening. Newer CT image acqui-
Response to chemotherapy is easier to evaluate sition techniques have been developed to facilitate
with objective decrease in tumor volume and absence 4-D imaging (time being the 4th dimension), and to
of progressive or new parenchymal and mediastinal decrease radiation dose using methods like iterative
disease. reconstruction (Sieren et al. 2010).
Size estimation can be done using unidimensional Dual energy CT scans are also being touted as
RECIST criteria or bidimensional WHO criteria. improving detection and characterizing the lung
Recent work has suggested that quantitative image lesions without significant change in radiation dose.
processing of serial CT scans acquired before and after Newer nodule detection software and rapidly
therapy administration may be a useful approach. advancing software algorithms for segmentation and
Complications which can occur secondary to 3-D volumetric evaluation of nodules and masses will
therapy are better imaged with CT especially che- further refine detection and treatment monitoring.
motherapy induced parenchymal toxicity. Dynamic and diffusion-weighted (DWI) MRI are
Very few reports in the literature exist on imaging in development and evaluation to improve specificity
for local recurrence. There is no good evidence-based in differentiating benign versus malignant nodules
data to support routine surveillance imaging (Walsh (Sieren et al. 2010).
et al. 1995) after therapy for local or distant recurrent As we further explore these new horizons, deter-
disease. However it is the accepted norm at most mine their clinical utility, aiming to develop safer and
centers and may have more relevance as more tailored more accurate form of imaging, one also hopes to see
surgical approaches become more common. Follow- progress in targeted imaging to complement the rap-
up imaging after therapy, which may or may not idly advancing tailored therapy, while we slowly
include chemotherapy, radiation therapy and surgery, unlock the secrets of lung cancer development.
Radiologic Imaging of Lung Cancer 73

Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D


References (2011) Global cancer Statistics. CA Cancer J Clin [Epub
ahead of print]
Johnson BE (1998) Second lung cancers in patients after
AJCC Cancer Staging Handbook - Springer Seventh Edition treatment for an initial lung cancer. J Natl Cancer Inst
(2010) 253–270 90(18):1335–1345
Akata S, Kajiwara N, Park J et al (2008) Evaluation of chest Klein JS, Braff S (2008a) Imaging evaluation of the solitary
wall invasion by lung cancer using respiratory dynamic pulmonary nodule. Clin Chest Med 29(1):15–38
MRI. J Med Imaging Radiat Oncol 52(1):36–39 Klein JS, Braff S (2008b) Imaging evaluation of the solitary
Aoki T, Nakata H, Watanabe H et al (2000) Evolution of pulmonary nodule. Clin Chest Med 29:15–38
peripheral lung adenocarcinomas CT findings correlated Kundel HL (1981) Predictive value and threshold of detect-
with histology and tumor doubling time. AJR Am J ability of lung tumors. Radiology 139:25–29
Roentgenol 174:763–768 Kuriyama K, Tateishi R, Doi O, Higashiyama M, Kodama K et al
Aoki T, Tomoda Y, Watanabe H (2001) Peripheral lung (1991) Prevalence of air bronchograms in small peripheral
adenocarcinoma: correlation of thin-section CT findings carcinomas of the lung on thin-section CT: comparison
with histologic prognostic factors and survival. Radiology with benign tumors. AJR, Am J Roentgenol 156:
220:803–809 921–924
Arita T, Matsumoto T, Kuramitsu T et al (1996) Is it possible to Lau CL, Harpole DH Jr (2000) Noninvasive clinical staging
differentiate malignant mediastinal nodes from benign nodes modalities for lung cancer. Semin Surg Oncol 18(2):
by size? Reevaluation by CT, transesophageal echocardiog- 116–123
raphy, and nodal specimen. Chest 110(4):1004–1008 Libby DM, Smith JP, Altorki NK et al (2004) Managing the
Awai K, Murao K, Ozawa A et al (2004) Pulmonary nodules at small pulmonary nodule discovered by CT. Chest
chest CT: effect of computer-aided diagnosis on radiolo- 125:1522–1529
gists’ detection performance. Radiology 230:347–352 Marom EM, McAdams HP, Erasmus JJ et al (1999) Staging
Bittner RC, Felix R (1998) Magnetic resonance (MR) imaging non–small cell lung cancer with whole-body PET. Radiol-
of the chest: state-of-the art. Eur Respir J 11:1392–1404 ogy 212:803–809
Cascade N, Gross BH, Kazerooni EA, Quint LE, Francis IR, Muhm JR, Miller WE, Fontana RS, Sanderson DR, Uhlenhopp
Strawderman M, Korobkin M (1998) Variability in the MA (1983) Lung cancer detected during a screening
detection of enlarged mediastinal lymph nodes in staging program using four month chest radiographs. Radiology
lung cancer: a comparison of contrast-enhanced and unen- 148:609–615
hanced CT. Am J Roentgenol 170:927–931 National cancer Institute. www.cancer.gov NSLT Lung cancer
Detterbeck FC, Jantz M, Wallace M et al (2007) Invasive trial results show mortality benefit with low-dose CT
mediastinal staging of lung cancer: ACCP evidence-based Twenty percent fewer lung cancer deaths seen among those
clinical practice guidelines (2nd edition). Chest 132(Suppl who were screened with low-dose spiral CT than with chest
3):202S–220S X-ray (Posted: 11/04/2010)
Erasmus JJ, Sabloff BS (2008) CT, positron emission tomog- O’Connor J, Jackson A, Asselin M, Buckley D, Parker G,
raphy, and MRI in staging lung cancer. Clin Chest Med Jayson G (2008) Quantitative imaging biomarkers in the
29(1):39–57 clinical development of targeted therapeutics: current and
Gorlova O, Peng B, Yankelevitz D, Henschke C, Kimmel M future perspectives. Lancet Oncol 9(8):766–776
(2005) Estimating the growth rates of primary lung tumours Padhani AR, Ollivier L (2001) The RECIST criteria: implica-
from samples with missing measurements. Stat Med tions for diagnostic radiologists. Br J Radiol 74:983–986
24(7):1117–1134, 4/2005. PMID: 15568189 Patz EF Jr, Erasmus JJ, McAdams HP et al (1999) Lung cancer
Gu P, Zhao YZ, Jiang LY et al (2009) Endobronchial staging and management: comparison of contrast-enhanced
ultrasound-guided transbronchial needle aspiration for stag- and nonenhanced helical CT of the thorax. Radiology
ing of lung cancer: a systematic review and metaanalysis. 212:56–60
Eur J Cancer 45:1389–1396 Primack SL, Lee KS, Logan PM et al (1994) Bronchogenic
Hansell DM, Lynch DA, Page McAdams H, Bankier AA carcinoma: utility of CT in the evaluation of patients with
(2010) Imaging of diseases of the chest, 5th edn. Elsevier suspected lesions. Radiology 193:795–800
publications, Philadelphia Ravenel JG, Mohammed TLH, Movsas B, Ginsburg ME et al
Heelan RT, Demas BE, Caravelli JF et al (1989) Superior (2010) ACR Appropriateness criterias noninvasive clinical
sulcus tumors: CT and MR imaging. Radiology 170(3 Pt 1): staging of bronchogenic carcinoma. J Thorac Imaging
637–641 25(4):W107–W111
Henschke CI, Yankelevitz DF, Mirtchev R et al (2002) The Revel M, Lefort C, Bissery A et al (2004) Pulmonary nodules:
ELCAP group, CT screening for lung cancer frequency and preliminary experience with three-dimensional evaluation.
significance of part-solid and nonsolid nodules. AJR Am J Radiology 231:459–466
Roentgenol 178:1053–1057 Rusch V et al (2009) The IASLC lung cancer staging project: a
Henschke CI, Boffetta P, Gorlova O, et al (2010) Assessment of proposal for a new international lymph node map in the
lung-cancer mortality reduction from CT screening. Lung forthcoming seventh edition of the TNM classification for
Cancer 16 (PubMed PMID:21168236) lung cancer. J Thorac Oncol 4(5):568–577
Hierholzer J, Luo L, Bittner RC et al (2000) MRI and CT in the Sieren JC, Ohno Y, Koyama H et al (2010) Recent technolog-
differential diagnosis of pleural disease. Chest 118:604–660 ical and application developments in computed tomography
74 P. Shah and J. L. Mulshine

and magnetic resonance imaging for improved pulmonary UyBico SJ, Wu CC, Suh RD, Le NH, Brown K, Krishnam MS
nodule detection and lung cancer staging. J Magn Reso- (2010) Lung cancer staging essentials: the new TNM
nance Imaging 32:1353–1369 Staging system and potential imaging pitfalls. Radiograph-
Simon GR, Turrisi A (2007) American College of Chest ics 30:1163–1181 doi:10.1148/rg.305095166
Physicians, Management of small cell lung cancer: ACCP van Klaveren RJ, Oudkerk M, Prokop M (2009) Management
evidence-based clinical practice guidelines (2nd edition). of lung nodules detected by volume CT scanning. N Engl J
Chest 132(3 Suppl):324S–339S Med 361:2221–2229
The National Lung Screening Trial: Overview and Study Vansteenkistel J, Dooms C, De Leyn P (2010) Early stage non-
Design Published online before print November 2, 2010, small-cell lung cancer: challenges in staging and adjuvant
doi:10.1148/radiol.10091808, January 2011 Radiology treatment: evidence-based staging. Ann Oncol 21(Supple-
258:243–253 ment 7):vii189–vii195
Toh CK (2009) The changing epidemiology of lung cancer. Walsh GL et al (1995) Is follow-up of lung cancer patients after
Methods Mol Biol 472:397–411 resection medically indicated and cost-effective? Ann
Travis WD, Brambilla E, Müller-Hermelink HK, Harris CC Thorac Surg 60(6):1563–1570 discussion 1570-2, 12/1995
(eds) (2004) World Health Organization lung, pleura, Yankelevitz DF, Henschke CI et al (1997) Does 2-year stability
thymus and heart. Pathology & Genetics tumours of the imply that pulmonary nodules are benign? AJR Am J
lung, pleura thymus and heart. IARC Press, Lyon Roentgenol 168(2):325–328
Travis WD, Brambilla E, Noguchi M, Nicholson AG (2011) Zerhouni EA, Stitik FP, Siegelman SS et al (1986) CT of the
International association for the study of lung cancer/ pulmonary nodule: a cooperative study. Radiology 160:319–327
american thoracic society/european respiratory society Zwirewich C, Vedal S, Miller R, Muller N (1991) Solitary
international multidisciplinary classification of lung adeno- pulmonary nodule:High-resolution CT and radiologic-
carcinoma. J Thorac Oncol 6(2):244–285 pathologic correlation. Radiology 179:469
PET/CT for Staging and Diagnosis
of Lung Cancer
Sigrid Stroobants

Contents Abstract
Positron emission tomography (PET) is an imaging
1 Basic Principles and Technical Improvement ..... 75 technique, which allows for accurate non-invasive
1.1 Tracers........................................................................ 76 measurements of metabolic pathways in tissues of
1.2 Conventional Versus Time-of-Flight (TOF)-PET .... 76
1.3 Technical Advances in Hybrid Systems................... 76
man in vivo. The most frequently used tracer in
PET oncology is the glucose analog 18F-fluoro-2-
2 Diagnosis of Solitary Pulmonary Nodules ............ 79
deoxy-glucose (FDG). The preferential accumulation
2.1 Strategies to Reduce the Number
of False Positives....................................................... 79 of FDG in neoplastic cells permits differentiation
2.2 Strategies to Reduce the Number between benign and malignant tissue. The ability to
of False Negatives ..................................................... 80 perform whole-body imaging within one examina-
3 Staging of Non-Small Cell Lung Cancer .............. 80 tion without increasing the radiation burden makes it
3.1 T-Staging.................................................................... 80 an ideal technique to ‘‘screen’’ patients for cancer
3.2 N-Staging ................................................................... 81 deposits. Also in thoracic oncology, FDG-PET has
3.3 M-Staging .................................................................. 81
proven its superiority over other imaging techniques
4 Small-Cell Lung Cancer ......................................... 83 in staging nodal and metastatic disease. However, the
5 Implementation in Clinical Practice ..................... 85 poor anatomic detail of PET can lead to errors in
References.......................................................................... 86
diagnosis and staging. Through the integration of
computer tomography (CT) and PET into one
machine, form and function are merged to create a
better imaging tool. In this chapter, we will highlight
the recent developments in hybrid machinery
(time-of-flight PET, PET-MR) and review the role
of integrated PET/CT in the diagnosis and staging of
lung cancer.

1 Basic Principles and Technical


Improvement

Positron emission tomography (PET) is a functional


S. Stroobants (&)
imaging modality which uses small amounts of phar-
Department of Nuclear Medicine,
University Hospital Antwerp, maceuticals labeled with positron-emitting radioiso-
Wilrijkstraat 10, 2650 Edegem, Belgium topes for metabolic imaging. Because of their similarity
e-mail: sigrid.stroobants@uza.be

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 75
DOI: 10.1007/174_2011_323, Ó Springer-Verlag Berlin Heidelberg 2011
76 S. Stroobants

to naturally occurring atoms in the human body, unstable. They decay by emission of a positron, which is
positron emittors as carbon-11 (11C), nitrogen-13 (13N), the subatomic, positively charged, antiparticle of the
and fluor-18 (18F) can be incorporated in biological negatively charged electron. A positron transverses a
molecules, without significantly influencing their short distance through the tissue (0.6 mm for 18F) until it
physiological and biochemical interactions. This allows combines with an electron in the surrounding media
in vivo imaging of metabolic pathways and receptor– (annihilation). This generates a pair of photons which
ligand interactions at pico-to-nanomolar sensitivities. travel in nearly opposite directions (180° apart) with an
energy of 511 keV each. These opposite photons can be
detected by detector pairs installed in a ring-shaped
1.1 Tracers
pattern in the PET camera. Photons that simultaneously
interact with these detectors are registered as decay
Although, there are numerous PET tracers described
events along a line-of-response (LOR). Based on these
in the literature, evidence of PET in oncology mainly
records, tomographic images of the regional radioac-
rests upon the glucose analog 18F-fluoro-deoxyglu-
tivity distribution are reconstructed (emission images).
cose (FDG), capitalizing that cancer has higher glu-
In conventional PET, the actual location where the
cose metabolism than most tissues (Pauwels et al.
annihilation occurred along the LOR is not measured,
1998). Since PET relies on the detection of metabolic
which inherently generates blurring in the reconstructed
alterations observed in cancer cells, this examination
image. The availability of faster detectors and electron-
yields data independently of associated structural
ics allows to measure the time difference between
characteristics. However, since FDG accumulation is
detection of each photon pair. In TOF-PET this addi-
not tumor specific but can also be present in inflam-
tional timing information is used to better localize the
mation, clinically relevant positive findings often
event within a small range along each LOR. The better
require confirmation.
localization of each event using TOF combined with
While FDG has made the way for PET in oncol-
more powerful reconstruction algorithms improves the
ogy, clinical practice with other tracers is limited. In
image quality especially in larger patients (Kadrmas
order to reduce tracer uptake in inflammatory tissue,
et al. 2009) (Fig. 1) . This leads to better lesion detection
more cancer-specific tracers were developed. The
and results in faster acquisition times which makes a
most promising one is probably 18F-fluoro-thymidine
typically whole-body PET/CT feasible in 10–15 min.
(FLT), a marker of cell proliferation with no or less
intense uptake in inflammatory tissue (Shields et al.
1998; Yap et al. 2006). In the study of Yang et al.
1.3 Technical Advances in Hybrid
(2010), FLT and FDG-PET were compared for pri-
Systems
mary staging of non-small cell lung cancer (NSCLC)
in 31 patients. FDG proved to be more sensitive for
Interpretation of PET scans is hampered by the lack of
detection of the primary tumor compared to FLT (94
anatomical detail which makes it sometimes difficult
vs. 74%) due to more intense trapping of the tracer
to correctly localize hot spots or differentiate tumor
(mean FDG-SUV = 7.7 vs. mean FLT-SUV = 4.2,
tissue from benign structures with physiological high-
p = 0.002). For nodal staging, FLT proved to be
FDG uptake as seen in muscle, brown fat, gut or
more specific (98 vs. 84%) but less sensitive (65 vs.
inflammation. These challenges are largely resolved
85%). Therefore, the role of FLT-PET for staging will
by the introduction of the combined PET/CT scanner.
be limited. More promising is its use for treatment
Current designs comprise a CT scanner in tandem
response evaluation, especially after targeted therapy.
with a PET scanner with a common patient bed for
both systems. Although the scanner appears exter-
1.2 Conventional Versus Time-of-Flight nally as a single device, internally there is little or no
(TOF)-PET mechanical integration. CT and PET images are
acquired consecutively for the same axial extent with
The biodistribution of the positron-emitting tracers is a simple horizontal translation of the bed. Upon
measured using a PET camera. Positron-emitting iso- completion of the scans, CT and PET images are
topes have an excess of protons and are, therefore, co-registered with fusion software and can be viewed
PET/CT for Staging and Diagnosis of Lung Cancer 77

Fig. 1 Principle of Time-of-Flight (TOF)–PET (a) which also compared to conventional PET. Example of the same patient
integrate the time difference by witch photon pairs are detected scanned twice with an interval of 3 months with conventional
in the reconstruction algorithm, resulting in less noisy images PET (b) and TOF-PET (c)

either separately with linked cursors or superimposed PET/CT protocol is used starting with a low-dose
with a selectable blending of the two modalities. non-enhanced CT (for attenuation correction of PET
An additional advantage of PET/CT is the marked data) followed by PET and than complemented
reduction in scan time (-50%) compared to with an appropriate contrast-enhanced CT protocol
PET-alone scanners since the CT images can also (De Wever et al. 2009). This could be of particular
be used to generate attenuation correction factors to importance when PET/CT is used for radiotherapy
be applied to the PET data to generate quantitative planning in patients with centrally located NSCLCs
images. CT-based attenuation correction can however given the complex anatomical setting (Pfannenberg
introduce specific artefacts due to patient motion or et al. 2007) (Fig. 3).
dense objects which then propagate into the PET The success of PET/CT in clinical practice and the
images (Mawlawi et al. 2006) (Fig. 2). Therefore, advantages of MRI compared to CT with respect to
non-attenuation-corrected PET images should also be radiation exposure and tissue characterization
reviewed to recognize these artefacts. have stimulated interest in the development of hybrid
Since the installation of the first clinical PET/CT in PET/MR imaging systems (Schlemmer et al. 2009).
2001, the technology has gained widespread use and The first strategy was to develop a sequential system:
all new PET scanners installed today are integrated a whole-body MR scanner and a PET scanner within
PET/CTs. The initial hybrid systems often contained the same or adjacent room operated by a single
only a single- or dual-slice CT scanner resulting in acquisition console and with a table that shuttles
inferior image quality of CT compared to stand-alone between the two systems. The advantage is that both
systems. Therefore, initial literature data mostly machines can still be used separately, but potential
included low-dose CT protocols without contrast misalignment due to patient motion is higher. The
enhancement. Modern scanners are however equipped design of a fully integrated system is more challeng-
with the latest generation 16–128 slice-MDCT, ing since conventional PET detectors are based on
therefore enabling the use of a true diagnostic CT as relatively bulky photomultipliers (PMTs). PMTs are
part of PET/CT. In this setting, often a multi-step very sensitive even to weak-magnetic fields and are
78 S. Stroobants

Fig. 2 Patient with extensive disease SCLC with multiple misalignment between PET and CT. Therefore, when no
liver and bone metastasis on PET (a, b). A liver metastasis in anatomical correlate is seen on CT (d), adjacent slices should
the liver dome is erroneously regarded as a lung metastasis of also be viewed to localize the lesion (e)
fusion images (c) due to a breathing artifact resulting in

Fig. 3 Patient with a NSCLC of the left hilum invading the mediastinum and with retro-obstructive atelectasis (c). The relation
of the tumor with the mediastinal structures is much better visualized on a contrast-enhanced CT (a) compared to low dose CT (b)

therefore not suitable to be used in a combined MR/ within an MRI scanner. Therefore alternative, new
PET scanner. In addition, its size would steal too semiconductor-based light detectors, such as ava-
much space from the open magnet bore if integrated lanche photodiodes (APDs) are replacing PMTs in
PET/CT for Staging and Diagnosis of Lung Cancer 79

integrated MR/PET. In spring 2011, the first inte- 97% (range 83–100) and a specificity of 78% (range
grated whole-body PET/MR was installed in Munich. 52–100) was obtained.
Workflows still need to be optimized (in contrast to
PET/CT, MR is now the most time-consuming part)
and indications which truly benefit from integrated 2.1 Strategies to Reduce the Number
PET/MR need to be defined. Given the high-equip- of False Positives
ment costs and lower-throughput capabilities com-
pared to PET/CT, widespread use of PET/MR in the False positives occur due to trapping of FDG in
near future is unlikely (Hicks and Lau 2009). activated granulocytes and/or macrophages in several
inflammatory conditions. Specificity across the
different studies is extremely variable depending on
2 Diagnosis of Solitary Pulmonary the prevalence of certain inflammatory or infectious
Nodules diseases. A nice pictorial overview of false-positive
findings is given in Shim et al. (2006a).
Solitary pulmonary nodules (SPNs) represent a Several efforts have been made to improve the
diagnostic challenge and with the increased use of specificity of PET. Since the uptake of FDG in benign
low-dose spiral CT for lung cancer screening, the lesions tend to be lower compared to that in malignant
number of coincidental SPNs will only increase (Tan tissue, quantification of the FDG uptake was used to
et al. 2003). The differential diagnosis of an SPN also improve the diagnostic accuracy. In the literature, a
includes inflammatory and infectious diseases and SUV above 2.5 is often used to discriminate benign
vascular, traumatic or congenital lesions besides can- from malignant nodules but without significant
cer. In most cases an histopathological proof is aimed increase in accuracy (meta-analysis, Gould et al.
for but this can be challenging in peripheral nodules. 2001). In fact, the use of a threshold value can
Moreover, in certain patients, who are at increased risk decrease the sensitivity in small lesions substantially
during invasive procedures or with lower probability compared to the simple visual analysis because of
for cancer, it may be desirable to further characterize a considerable underestimation of the true activity due
pulmonary nodule by imaging, rather than proceeding to partial volume effects, through which the SUV
immediately to tissue diagnosis. measurement drops under the threshold, although the
Before the PET era, CT was the principal imaging lesion is clearly visible. In the study of Lowe et al.
modality to evaluate indeterminate nodules. Charac- (1994), the sensitivity for detecting malignant nodules
terization is based on the shape, borders, density and \1.5 cm decreased from 100% using visual analysis
contrast enhancement (Siegelman et al. 1986; to 80% using the threshold SUV value of 2.5.
Swensen et al. 2000). Lesions are usually considered Not only the amount of FDG uptake but also the
benign if they show the following features: concentric tracer kinetics are thought to be different in benign
calcifications, round shape or a morphologic stability and malignant tissue, with continuous uptake in
over 2 years (Yankelevitz and Henschke 1997). malignant lesions and a rapid uptake followed by a
On the contrary, malignant features typically include fast and then gradual washout in benign masses.
ill-defined margins, spiculation, cavitation, invasion Using dual time point imaging at 1 and 2 h after
of bronchi or vessels and a doubling time of tracer injection, an increase of at least 10% in SUV
\10 months (Gurney et al. 1993). CT has an excellent between the first and the second scan proved to
sensitivity (96%, range 91–98%) for detection of SPN be more accurate than a SUV threshold of 2.5
but a poor specificity (50%, range 41–58%) (Swensen (Alkhawaldeh et al. 2008). Unfortunately, dual time
et al. 2003). In the past years, FDG-PET has been imaging does not increase specificity in granuloma-
studied extensively for characterization of SPNs and tous diseases like sarcoidosis or tuberculosis
multiple studies have showed that for nodules [1 cm (Sathekge et al. 2010) since FDG uptake tends to
PET had similar sensitivity but superior specificity as increase over time, similar to cancer.
compared to CT (Gould et al. 2001; Fischer et al. Instead of a binary reading (positive or negative),
2001; Ung et al. 2007). In the meta-analysis of Gould maybe it is more appropriate to report the PET result as
et al. (2001) in 1,474 lesions, an overall sensitivity of a probability of cancer. A large prospective series
80 S. Stroobants

(n = 585) looked at the accuracy of integrated PET/CT (Ashraf et al. 2011). Twenty of the 54 nodules proved to
scan in SPNs B2.5 cm (Bryant and Cerfolio 2006). If be malignant (mean diameter 13 mm). In a multivari-
the SUVmax was between 0 and 2.5 there was a 24% able analysis, both FDG uptake (equal to or greater than
chance of malignancy, if between 2.6 and 4.0 it was the mediastinal blood pool) and volume doubling time
80%, and if[4 it was 96%. It may however be neces- (VDT \ 1 year) were independently associated with
sary to define the SUV ranges for given probabilities for malignancy but the combination of both was better for
each institution individually because of differences in predicting lung cancer than either procedure alone. The
the patient population and also because the magnitude highest sensitivity (90%) was obtained when either
of calculated SUV for the same lesion can vary PET or VDT indicated malignancy.
depending on the PET system and method of attenua-
tion correction utilized (Boellaard 2009).
3 Staging of Non-Small Cell Lung
Cancer
2.2 Strategies to Reduce the Number
of False Negatives Staging of NSCLC is done according to the tumor (T),
node (N), metastasis (M) system.
For detection on PET a critical mass of metabolically Recently the classification was updated (7th edi-
active malignant cells is required. False-negative find- tion) and introduces new dimension ranges for T and
ings therefore occur in tumors with low-metabolic a different interpretation of additional tumor nodules,
activity like carcinoid tumors (Belhocine et al. 2002) or pericardial and pleural involvement, with consequent
tumors with low-cell density like mucinous tumors variations in the definition of tumor stages (Goldstraw
(Berger et al. 2000) or the alveolar subtype of bron- et al. 2007; Goldstraw 2009). Currently all the liter-
choalveolar carcninoma (BAC) (Heyneman and Patz ature data on impact of PET on TNM stage is based
2002). Nomori et al. (2004) evaluated the performance on the previous classification (Mountain 1997).
of PET in 15 patients with ground glass opacities, a
typical CT pattern for non-invasive BAC. PET was
only positive in 1/10 malignant lesions and false posi- 3.1 T-Staging
tive in 4/5 benign lesions (focal pneumonia), resulting
in a sensitivity of 10% and a specificity of 20%. The assessment of the primary tumor extension is
The sensitivity of PET is also lower in sub-centi- usually based on thoracic CT, occasionally comple-
metric lesions. One study evaluated the performance of mented by MRI. Due to the increased image quality of
PET in 136 uncalcified nodules \3 cm. All of the 20 MDCT, scanners can depict with greater confidence an
lesions \1 cm were negative on PET, eight of who invasion of a tumor in surrounding tissues by assess-
were malignant (Nomori et al. 2004). Recently, three ment of preserved mediastinal fat planes and can detect
studies have reported on the value of selected PET as a more and smaller lesions (Verschakelen et al. 2004).
second-step test in lung cancer screening. Pastorino PET on itself does not add much to the assessment of
et al. (2003) applied PET on non-calcified nodules local resectability because its inferior spatial resolution
C7 mm. FDG-PET was positive in 18/20 cancers. One does not give more detail of the exact tumor extent or
8 mm adenocarcinoma and one 11 mm predominantly infiltration of neighboring structures. In recent studies
BAC were missed. In the study by Bastarrika et al. using PET/CT, it was therefore not surprising that T
(2005), PET was applied on nodules [10 mm or stage was more accurately assessed on PET/CT
growing nodules C7 mm, with a sensitivity and nega- compared to PET alone (Lardinois et al. 2003; Antoch
tive predictive value of 69 and 71%, respectively. By et al. 2003; Cerfolio et al. 2004; Shim et al. 2005; De
including a 3-month follow-up CT after a negative Wever et al. 2007a). In a minority of the patients, PET/
PET, the authors were able to increase sensitivity and CT also proved to be superior to CT alone due to a
negative predictive value to 100%. Finally, patients better discrimination between the tumor and sur-
with indeterminate nodules included in the Danish rounding atelectasis or inflammation and to correct
Lung Cancer Screening Trial referred for a 3-month exclusion of tumor involvement in co-existing lung
re-scan and PET were retrospectively analyzed nodule(s) in the same lobe (De Wever et al. 2009).
PET/CT for Staging and Diagnosis of Lung Cancer 81

Some studies also describe an additional value of spots, therefore it is not surprising that recent studies
PET for detection of pleural metastasis. In a larger comparing integrated PET/CT vs. CT alone or PET
study in 92 patients with pleural effusion, of whom alone show a higher accuracy for the hybrid technique
71% were deemed indeterminate on CT, PET had a (De Wever et al. 2009).
sensitivity, specificity, and accuracy of 100, 71 and While spatial resolution of PET/CT has improved,
80%, respectively (Schaffler et al. 2004). The speci- the techniques remain inadequate to rule out sub-cen-
ficity and positive predictive value (PPV) were lower timeter tumor deposits (Ikeda et al. 2006). Billè et al.
than described previously, due to the larger number of (2009) evaluated the likelihood of finding mediastinal
benign pleural effusions. One study specifically N2 disease after a negative PET/CT scan in a con-
looked at the value of PET in dry pleural dissemina- secutive cohort of 159 patients. Factors associated with
tion. Since this is often caused by multiple very small unforsceen N2 were central tumors, adenocarcinoma
pleural nodules beyond the resolution of a PET sys- histology, PET/CT hilar N1 disease and CT mediasti-
tem, the sensitivity was only 25% (Shim et al. 2006b). nal N2 disease. The latter was also recognized in the
meta-analysis of Langen et al. (2006) where a negative
PET scan resulted in a post-PET probability for N2
3.2 N-Staging disease of 5% for LN \ 15 mm, compared to 21% for
LN C 16 mm. These data suggest that these patients
The accuracy of CT for the prediction of intrathoracic should be referred for invasive surgical staging prior to
nodal spread of tumors remains limited and the more possible thoracotomy to prevent too many unnecessary
recently developed CT systems do not change this thoracotomies in this subset.
because nodal staging with CT is only based on size The PPV of PET is less optimal and therefore tissue
criteria. The current consensus considers a node confirmation of PET-positive nodes is always manda-
\10 mm in short axis diameter suspect for metastatic tory to avoid denial of radical surgery based on
involvement, which results in sensitivity ranging from 52 false-positive findings. In the early PET studies, a
to 69% and specificity 69 to 82% (Dillemans et al. 1994). lymph node was reported positive when FDG uptake
Over the past years, several studies have found that was more intense than the surrounding background.
FDG-PET has a significantly higher sensitivity and False positives were related to the presence of inflam-
specificity than CT for mediastinal staging and matory conditions. The introduction of PET/CT and
this superiority has been confirmed in different TOF-PET however improved the sensitivity resulting in
meta-analyses (Fischer et al. 2001; Gould et al. 2003; visualization of ‘‘normal’’ nodes above the mediastinal
Silvestri et al. 2007). In the most recent one of Silvestri background. In the study of Lee et al. (2007), the PPV of
et al. data from 44 studies including 2,865 patients, PET/CT was only 56% compared to 68% with
resulted in a pooled estimates of sensitivity and stand-alone PET in a comparable historical cohort.
specificity for identifying mediastinal metastasis of 74 The drastic increase in false-positive results reinforced
(95% CI, 69–79%) and 85% (95% CI, 82–88%), the need for surgical staging. Further studies will need to
respectively. Corresponding positive and negative clarify how PET-positive nodes should be defined with
likelihood ratios for mediastinal staging with PET TOF-PET-CT to achieve a better accuracy.
scanning were 4.9 and 0.3, respectively. These findings
demonstrate that PET scanning is more accurate than CT
scanning for staging of the mediastinum in patients with 3.3 M-Staging
lung cancer, though it is far from perfect.
The superiority of FDG-PET is explained by the more The observation of metastases in patients with
frequent correct identification of ‘‘small malignant NSCLC implies that a patient can no longer be cured.
nodes’’ and ‘‘large benign nodes’’. Errors are related to Forty percent of the patients with NSCLC have dis-
incorrect localization, minimal tumor load (false nega- tant metastases at presentation, most commonly in the
tives) and inflammation on (false positives) (Fig. 4). adrenal glands, bones, liver or brain (Quint et al.
Due to the lack of anatomical information in the 1996). The current standard non-invasive staging tests
PET image, reading of PET scan in the presence of (including ultrasound, CT, MRI and bone scintigra-
the CT images is crucial for correct localization of hot phy) are far from perfect. A systemic relapse develops
82 S. Stroobants

Fig. 4 Patient with adenocarcinoma of the right upper lobe underwent thoracotomy. Final stage was pT2aN1M0. This case
and with FDG avid lymph nodes in the right hilum (a) and underlines the importance to always confirm PET positives
subcarinal region (b, c). Metastatic involvement could not be nodes with histology
confirmed with EBUS nor mediastinoscopy so patient

in up to 20% of surgically treated patients in the chances for radical therapy. In these patients, a con-
period from 3 to 24 months after complete surgical firmatory test is indicated. Accurate localization of
resection (Pantel et al. 1996). FDG-avid regions on fused PET/CT images reduces the
PET identifies metastatic spread in 5–29% of risk of false-positive interpretations of physiological
patients with negative or equivocal conventional phenomena such as uptake in bowel or metabolically
imaging (Schrevens et al. 2004). In recent studies using active brown fat. Accordingly, previous estimates of
integrated PET/CT, the hybrid modality was signifi- the impact of PET on the management of lung cancer
cantly better than CT or PET alone for extra thoracic are likely to be surpassed in current clinical practice.
metastases, although it is limited in assessing brain Enlarged adrenal glands on CT are found in up to
metastases (De Wever et al. 2007b). While some PET 20% of NSCLC patients, but up to two-thirds of these
images can be considered definite proof of multi-focal lesions are benign adenomas. PET is reported to have
metastatic disease, caution is always indicated in soli- a high sensitivity (reaching 100%) in the detection of
tary extrathoracic PET findings that determine the adrenal metastases, which means that an equivocal
PET/CT for Staging and Diagnosis of Lung Cancer 83

lesion of [1 cm on CT without FDG uptake on PET surrounding brain tissue. MRI (or CT) remains the
will usually not be metastatic. Specificity of PET for method of choice to stage the brain.
adrenal metastases is also high (between 80 and
100%), due to weak FDG uptake in adenomas.
Attempts are made to improve the specificity by using 4 Small-Cell Lung Cancer
SUV thresholds or adrenal/liver uptake ratios.
Recently, Brady et al. (2009) proposed an algorithm Small-Cell Lung Cancer (SCLC) represents only
combining density measurement on non-CE (HU [ 15–20% of all lung cancers, and is often dissiminated
10) and a SUV threshold (SUV [ 3.1) to define at the time of diagnosis, thereby obviating the need
malignancy. The proposed algorithm for PET/CT for PET in many patients. In contrast to NSCLC, a
reading proved to be more specific than PET or CT two-stage classification scheme is routinely used to
alone (86 vs. 76 vs. 60% respectively) without loss in define the extent of disease, which divides SCLC into
sensitivity (97%) (Fig. 5). limited disease (LD) and extensive disease (ED). LD
PET scanning appears to have excellent perfor- is defined as disease confined to one hemithorax, the
mance characteristics in assessing bone metastases, mediastinum and the supraclavicular lymph nodes.
with specificity, sensitivity, NPV, PPV and accuracy all All other patients are classified as having ED,
exceeding 90%, though false-positive and false- including those with malignant pleural effusion
negative findings are occasionally seen (Silvestri et al. (Rodriguez and Lilenbaum 2010). Diagnostic proce-
2007). The accuracy of PET scanning surpassed that of dures commonly used to stage the disease include
radionuclide bone scanning in two direct comparative chest and abdomen CT, brain CT or MRI, radionu-
studies (Hsia et al. 2002; Schirrmeister et al. 2004). clide bone scans and bone marrow aspiration.
Caution is however required with distal lesions The value of PET in staging SCLC has been
(e.g. below the knee), which will fall outside the field- evaluated by relatively small, mostly retrospective
of-view of a standard ‘whole-body’ PET acquisition, or studies, all indicating a possible role for PET (Ung
in osteoblastic lesions which are more readily seen on et al. 2007). PET seems to be especially promising in
Tc-99 m bone scan, as demonstrated in a study on breast the detection bone metastasis and supraclavicular
cancer patients (Cook et al. 1998). Because most bone nodes (Fig. 6). CT and MRI outperform FDG-PET
lesions in NSCLC are in the central skeleton, and nearly with respect to the detection of brain metastases.
all are osteolytic, PET scan usually replaces bone scan, Unlike in NSCLC, studies comparing PET for medi-
except in specific clinical indications. astinal staging with histopathology results are lacking.
For liver metastases, PET will have an additional Probably the accuracy is comparable to that of
value to conventional imaging because of its ability to NSCLC since in many of these studies, final histology
differentiate indeterminate hepatic lesions. There are proved to be SCLC in some patients, without higher
no specific series on the use of PET in patients with rate of false-positive or -negative results.
liver metastases from NSCLC. Some general series on In the systematic meta-analysis (Ung et al. 2007),
staging NSCLC suggest a superiority of PET by being sensitivity and specificity of PET for staging exten-
more accurate than CT comparable to other tumor sive versus limited-stage disease ranged between
types. In a meta-analysis comparing different imaging 89–100 and 78–95%, respectively. More recent
modalities in the detection of colorectal liver metas- studies confirm these data with change in stage clas-
tasis, helical CT, MR imaging at 1.5T and FDG-PET sification in 8–17% of the patients. In the study of
had similar sensitivities on a per-lesion analysis Vinjamuri et al. (2008), additional sites of disease
(Bipat et al. 2005). In routine clinical practice, CT were detected in 13 of 42 patients (32%), mostly
therefore remains the standard imaging technique located in bones or supraclavicular nodes. PET
for the liver. The use of PET is mainly to provide resulted in a change of disease stage in 16%
additional information for the differentiation of (upstaging in 4%, downstaging in 12%). Niho et al.
hepatic lesions that are indeterminate on conventional (2007) explored the additional value of PET in 63
imaging. patients with LD after conventional workup. ED was
FDG-PET is not sensitive enough to exclude brain found in 6/63 patients and additional LN involvement
metastases, due to the high-glucose uptake of normal was found 14%. In a recent study Azad et al. (2010) in
84 S. Stroobants

Fig. 5 Patient with a NSCLC of the right upper lobe and an metastases. In the latter, adrenal uptake is in mostcases clearly
indeterminate mass in the left adrenal gland (CT density = 24 higher than normal liver uptake. In this case, the diagnosis of
HU). PET only showed faint uptake in the adrenal mass (arrow) adenoma was confirmed with biopsy
with an SUV=2.4 suggestive for adenoma rather than

120 SCLC patients, PET up-staged 10 patients and While the results of PET staging are promising, we
down-staged three patients. Overall PET data resulted need larger prospective trials before definite conclu-
in a change of stage in 12% of patients. Fischer et al. sions can be drawn on the exact role of PET in the
(2007) report on the first prospective study using staging SCLC. The reference standard to which PET
PET/CT in 29 patients with SCLC. PET/CT proved to compared is variable among the studies, and none of
have a higher sensitivity than conventional workup the studies confirmed all lesions with histologic
(93 vs. 73%) with equal specificity (100%). In their results. Future studies should fully report the fre-
population PET/CT findings determined a change of quency of correct and incorrect staging changes when
stage in five of 29 patients (17%). PET is added to conventional tests and link diagnostic
PET/CT for Staging and Diagnosis of Lung Cancer 85

Fig. 6 Patient with SCLC of the left hilum with bulky limited to extensive disease was confirmed by biopsy of the hot
mediastinal involvement (a). PET also revealed hot spots in spot of lesion in the left sacrum (d)
the bone (b) which were not visible on CT (c). Upstaging from

performance to outcomes such as improvement in and surgical technique and one cannot give an
survival or reduced morbidity. Currently only Azad overall recommendation for ‘optimal’ diagnostic
et al. (2010) correlated PET stage with survival and workup since the optimal strategy will depend on the
found a longer OS in patients with ED on pre-PET availability and expertise of the different techniques
staging but downstaged to LD after PET, compared to as well as specific patient characteristics.
those with ED on PET (median 10.9 vs. 5.9 months; Although PET is the imaging technique with the
log-rank p = 0.037). highest accuracy for characterization of indeterminate
lung lesions, careful selection of patients taking into
account the likelihood of malignancy is important.
5 Implementation in Clinical Practice Patients with rapidly growing hypermetabolic nodules
outside areas of endemic tuberculosis/mucosis, should
Non-invasive lung cancer staging is substantially be referred immediately for surgical resection if the
improved by the use of PET. The most exciting lesion is easily accessable (VATS) and there is no
feature of PET is that it gives a reasonably cancer- contraindications for surgery. For patients with nodules
specific imaging of the entire patient in one single that are not FDG avid and have a long-doubling time, a
non-invasive test with a better accuracy than con- wait and see policy is acceptable. Lastly, for patients
ventional imaging, thus with a potential impact on with discordant findings on FDG-PET and volumetric
stage designation and therapeutic decision. Diagnosis analysis,the likelihood of cancer is intermediate to high
and staging of lung cancer has become a truly multi- requiring additional tissue sampling or strict FU (repeat
disciplinary process involving imaging, endoscopic CT within 3 months) (Gould et al. 2011).
86 S. Stroobants

When PET/CT is available, it is probably the best Bastarrika G, Garcia-Velloso MJ, Lozano MD et al (2005)
single test to stage a lung-cancer patients. The most Early lung cancer detection using spiral computed tomog-
raphy and positron emission tomography. Am J Respir Crit
added value is in the detection of distant metastasis Care Med 171:1378–1383
since this implies that the patient is no longer curable. Belhocine T, Foidart J, Rigo P et al (2002) Fluorodeoxyglucose
While some PET/CT images can be considered defi- positron emission tomography and somatostatin receptor
nite proof of multi-focal metastatic disease, caution is scintigraphy for diagnosing and staging carcinoid tumours:
correlations with the pathological indexes p53 and Ki-67.
always indicated in solitary extrathoracic findings that Nucl Med Commun 23:727–734
determine the chances for radical therapy. In these Berger KL, Nicholson SA, Dehdashti F et al (2000) FDG PET
patients, a confirmatory test is indicated. evaluation of muci-nous neoplasms: correlation of FDG
PET/CT is also useful for mediastinal staging but uptake with histopathologic features. Am J Roentgenol 174:
1005–1008
the clinical impact is less pronounced than initially Billè A, Pelosi E, Skanjeti A et al (2009) Preoperative
anticipated. The high-negative predictive value of intrathoracic lymph node staging in patients with non-
PET creates the possibility to omit invasive staging if small-cell lung cancer: accuracy of integrated positron
PET suggests absence of LN disease. However one emission tomography and computed tomography. Eur J
Cardiothorac Surg 36:440–445
should be very careful for false-negative PET findings Bipat S, van Leeuwen MS, Comans EF et al (2005) Colorectal
in particular situations. The spatial resolution of PET/ liver metastases: CT, MR imaging, and PET for diagnosis—
CT remains inadequate to rule out sub-centimeter meta-analysis. Radiology 237:123–131
lymph node metastasis and does not obviate the need Boellaard R (2009) Standards for PET image acquisition
and quantitative data analysis. J Nucl Med 50(Suppl 1):
for invasive staging procedures in patients with a 11S–20S
higher likelihood of mediastinal lymph node Brady MJ, Thomas J, Wong TZ et al (2009) Adrenal nodules at
involvement such as patients with central tumors, FDG PET/CT in patients known to have or suspected of
low-FDG uptake in the primary tumor, PET/CT hilar having lung cancer: a proposal for an efficient diagnostic
algorithm. Radiology 250:523–530
N1 disease and CT mediastinal N2 disease. If these Bryant AS, Cerfolio RJ (2006) The maximum standardized
rules of interpretation are handled correctly, relevant uptake values on integrated FDG-PET/CT is useful in
LN disease will rarely be missed. Minimal N2-disease differentiating benign from malignant pulmonary nodules.
may be discovered at surgical exploration, but Ann Thorac Surg 82:1016–1020
Cerfolio RJ, Ojha B, Bryant AS et al (2004) The accuracy of
resection in these patients is rewarding. False-positive integrated PET-CT compared with dedicated PET alone for
findings are due to the fact that FDG uptake is not the staging of patients with non small cell lung cancer. Ann
tumor specific, and can be found in all active tissues Thorac Surg 78:1017–1023
with high-glucose metabolism, in particular inflam- Cook GJ, Houston S, Rubens R, Maisey MN et al (1998)
Detection of bone metastases in breast cancer by 18FDG
mation. Therefore, clinically relevant FDG-avid PET: differing metabolic activity in osteoblastic and
mediastinal LNs should always be examined with the osteolytic lesions. J Clin Oncol 16:3375–3379
most appropriate tissue sampling technique. De Wever W, Ceyssens S, Mortelmans L et al (2007a)
Additional value of PET-CT in the staging of lung cancer:
comparison with CT alone, PET alone and visual correlation
of PET and CT. Eur Radiol 17:23–32
References De Wever W, Vankan Y, Stroobants S et al (2007b) Detection
of extrapulmonary lesions with integrated PET/CT in the
staging of lung cancer. Eur Respir J 29:995–1002
Alkhawaldeh K, Bural G, Kumar R, Alavi A (2008) Impact of De Wever W, Stroobants S, Coolen J et al (2009) Integrated
dual-time-point (18)F-FDG PET imaging and partial vol- PET/CT in the staging of non small cell lung cancer:
ume correction in the assessment of solitary pulmonary technical aspects and clinical integration. Eur Respir J
nodules. Eur J Nucl Med Mol Imaging 35:246–252 33:201–212
Antoch G, Stattaus J, Nemat AT et al (2003) Non-small cell Dillemans B, Deneffe G, Verschakelen J et al (1994) Value of
lung cancer: dual-modality PET/CT in preoperative staging. computed tomography and mediastinoscopy in preoperative
Radiology 229:526–533 evaluation of mediastinal nodes in non-small cell lung
Ashraf H, Mortensend J, Dirksen A et al (2011) Combined use cancer. A study of 569 patients. Eur J Cardiothorac Surg
of PET and volume doubling time in lung cancer screening 8:37–42
with low dose CT. Thorax 66:315–319 Fischer BM, Mortensen J, Hojgaard L (2001) Positron emission
Azad A, Chionh F, Scott AM et al (2010) High impact of 18F- tomography in the diagnosis and staging of lung cancer: a
FDGPET onmamagement and prognostic stratification of systematic, quantitative review. Lancet Oncol 2:659–666
newly diagnosed small cell lung cancer. Mol Imaging Biol Fischer BM, Mortensen J, Langer SW et al (2007) A
12:433–451 prospective study of PET/CT in initial staging of small-cell
PET/CT for Staging and Diagnosis of Lung Cancer 87

lung cancer: comparison with CT, bone scintigraphy and Niho S, Fujii H, Murakami K et al (2007) Detection of
bone marrow analysis. Ann Oncol 18:338–345 unsuspected distant metastases and/or regional nodes by
Goldstraw P (2009) The 7th edition of TNM in lung cancer: FDG-PET in LD-SCLC scan in apparent limited-disease
what now? J Thorac Oncol 4:671–673 small-cell lung cancer. Lung Cancer 57:328–333
Goldstraw P, Crowley J, Chansky K et al (2007) The IASLC Nomori H, Watanabe K, Ohtsuka T et al (2004) Valuation of
lung cancer staging project: proposals for the revision of the F-18 fluorodeoxyglucose (FDG) PET scanning for pulmon-
TNM stage groupings in the forth-coming (seventh) edition ary nodules less than 3 cm in diameter, with special
of the TNM classification of malignant tumours. J Thorac reference to the CT images. Lung Cancer 45:19–27
Oncol 2:706–714 Pantel K, Izbicki J, Passlick B et al (1996) Frequency and
Gould MK (2011) Evaluation of screening-detected lung prognostic significance of isolated tumour cells in bone
nodules: minimising the risk of unnecessary biopsy and marrow of patients with non-small cell lung cancer without
surgery. Thorax 66:277–279 overt metastases. Lancet 347:649–653
Gould MK, Maclean CC, Kuschner WG et al (2001) Accuracy Pastorino U, Bellomi M, Landoni C et al (2003) Early lung
of positron emission tomography for diagnosis of pulmon- cancer detection with spiral CT and positron emission
ary nodules and mass lesions: a meta-analysis. JAMA tomography in heavy smokers: 2-year results. Lancet 362:
285:914–924 593–597
Gould MK, Kuschner WG, Rydzak CE et al (2003) Test Pauwels EK, McCready VR, Stoot JH, van Deurzen DF (1998)
performance of positron emission tomography and com- The mechanism of accumulation of tumour-localising
puted tomography for mediastinal staging in patients with radiopharmaceuticals. Eur J Nucl Med 25:277–305
non-small cell lung cancer: a meta-analysis. Ann Intern Med Pfannenberg AC, Aschoff P, Brechtel K et al (2007) Low dose
139:879–892 non-enhanced CT versus standard dose contrast-enhanced
Gurney JW, Lyddon DM, McKay JA (1993) Determining the CT in combined PET/CT protocols for staging and therapy
likelihood of malignancy in solitary pulmonary nodules planning in non-small cell lung cancer. Eur J Nucl Med Mol
with Bayesian analysis. Part II. Application. Radiology Imaging 34:36–44
186:415–422 Quint LE, Tummala S, Brisson LJ et al (1996) Distribution of
Heyneman LE, Patz EF (2002) PET imaging in patients distant metastases from newly diagnosed non- small cell
with bronchioloalveolar cell carcinoma. Lung Cancer 38: lung cancer. Ann Thorac Surg 62:246–250
261–266 Rodriguez E, Lilenbaum RC (2010) Small cell lung cancer:
Hicks RJ, Lau EWF (2009) PET/MRI: a different spin from past, present and future. Curr Oncol Rep 12:327–334
under the rim. Eur J Nucl Med Mol Imaging 36(Suppl Sathekge MM, Maes A, Pottel H et al (2010) Dual time-point
1):S10–S14 FDG PET-CT for differentiating benign from malignant
Hsia TC, Shen YY, Yen RF et al (2002) Comparing whole body solitary pulmonary nodules in a TB endemic area. S Afr
18F-2-deoxyglucose positron emission tomography and Med J 100:598–601
technetium-99m methylene diophosphate bone scan to Schaffler GJ, Wolf G, Schoellnast H et al (2004) Non-small cell
detect bone metastases in patients with non-small cell lung lung cancer: evaluation of pleural abnormalities on CT
cancer. Neoplasma 49:267–271 scans with 18F FDG PET. Radiology 231:858–865
Ikeda K, Nomori H, Mori T et al (2006) Size of metastatic and Schirrmeister H, Arslandemir C, Glatting G et al (2004)
nonmetastatic mediastinal lymph nodes in non-small cell Omission of bone scanning according to staging guidelines
lung cancer. J Thorac Oncol 1:949–952 leads to futile therapy in non-small cell lung cancer. Eur J
Kadrmas DJ, Casey ME, Conti M et al (2009) Impact of Nucl Med Mol Imaging 31:964–968
time-of-flight on PET tumor detection. J Nucl Med 50: Schlemmer HP, Pichler BJ, Krieg R et al (2009) An integrated
1315–1323 MR/PET system: prospective applications. Abdom Imaging
Langen AJ, Raijmakers P, Riphagen I et al (2006) The size of 34:668–674
mediastinal lymph nodes and its relation with metastatic Schrevens L, Lorent N, Dooms C et al (2004) The role of PET
involvement: a meta-analysis. Eur J Cardiothor Surg 29:26–29 scan in diagnosis, staging, and management of non-small
Lardinois D, Weder W, Hany TF et al (2003) Staging of non- cell lung cancer. Oncologist 9:633–643
small-cell lung cancernwith integrated positron-emission Shields AF, Grierson JR, Dohmen BM et al (1998) Imaging
tomography and computed tomography. N Engl J Med proliferation in vivo with [F-18]FLT and positron emission
348:2500–2507 tomography. Nat Med 4:1334–1336
Lee BE, von Haag D, Lown T et al (2007) Advances in positron Shim SS, Lee KS, Kim BT et al (2005) Non-small cell lung
emission tomography technology have increased the need cancer: prospective comparison of integrated FDG PET-CT
for surgical staging in non-small cell lung cancer. J Thorac and CT alone for preoperative staging. Radiology 236:
Cardiovasc Surg 133:746–752 1011–1019
Lowe VJ, Hoffman JM, DeLong DM et al (1994) Semiquan- Shim SS, Lee KS, Kim BT et al (2006a) Focal parenchymal
titative and visual analysis of FDG-PET images in lung lesions showing a potential of false-positive and false-
pulmonary abnormalities. J Nucl Med 35:1771–1776 negative interpretations on integrated PET/CT. AJR Am J
Mawlawi O, Pan T, Macapinlac HA (2006) PET/CT imaging Roentgenol 186(2):639–648
techniques, considerations, and artifacts. Thorac Imaging Shim SS, Lee KS, Kim BT et al (2006b) Integrated PET/CT
21:99–110 and the dry pleural dissemination of peripheral adenocar-
Mountain CF (1997) Revisions in the international system for cinoma of the lung: diagnostic implications. J Comput
staging lung cancer. Chest 111:1710–1717 Assist Tomogr 30:70–76
88 S. Stroobants

Siegelman SS, Khouri N, Leo FP, Fishman EK et al (1986) Solitary Verschakelen JA, De Wever W, Bogaert J (2004) Role of
pulmonary nodules: CT assessment. Radiology 160:307–312 computed tomography in lung cancer staging. Curr Opin
Silvestri GA, Gould MK, Margolis ML et al (2007) Noninva- Pulm Med 10:248–255
sive staging of non-small cell lung cancer: ACCP Vinjamuri M, Craig M, Campbell-Fontaine A et al (2008) Can
evidenced-based clinical practice guidelines 132(Suppl 3): positron emission tomography be used as a staging tool for
178S–201S (2nd ed) small-cell lung cancer? Clin Lung Cancer 9:30–34
Swensen SJ, Viggiano RW, Midthum DE et al (2000) Lung Yang W, Zhang Y, Fu Z et al (2010) Imaging of proliferation
nodule enhancement at CT: multicenter study. Radiology with 18F-FLT PET/CT versus 18F-FDG PET/CT in non-
214:73–80 small-cell lung cancer. Eur J Nucl Med Mol Imaging
Swensen SJ, Jett JR, Hartman TE et al (2003) Lung cancer 37:1291–1299
screening with CT: Mayo clinic experience. Radiology Yankelevitz DF, Henschke CI (1997) Does 2-year stability
226:756–761 imply that pulmonary nodules are benign? Am J Roentgenol
Tan BB, Flaherty KR, Kazerooni EA et al (2003) The solitary 168:325–328
pulmonary nodule. Chest 123(Suppl 1):89S–96S Yap CS, Czernin J, Fishbein MC et al (2006) Evaluation of
Ung YC, Maziak DE, Vanderveen JA et al (2007) 18F- thoracic tumors with 18F-fluorothymidine and 18F-fluoro-
fluorodeoxyglucose positron emission tomography in the deoxyglucose-positron emission tomography. Chest 129:
diagnosis and staging of lung cancer: a systematic review. 393–401
J Natl Cancer Inst 99:1753–1767
Surgical Staging of Lung Cancer
for Advances in Radiation Oncology
of Lung Cancer
Farhood Farjah and Valerie W. Rusch

Contents Abstract
This chapter reviews surgical approaches to deter-
1 Introduction.............................................................. 89 mining T, N, and M status for lung cancer; an
2 Stage Classification and Determination ................ 90 expanded description of individual mediastinal
2.1 T-Stage....................................................................... 90 staging modalities; the evidence for invasive
2.2 N-Stage ...................................................................... 90 mediastinal staging procedures and multi-modality
2.3 M-Stage...................................................................... 92
staging strategies; and staging practices in the
3 Invasive Mediastinal Staging Modalities .............. 93 community at large.
3.1 Transthoracic Needle Biopsy .................................... 93
3.2 Transbronchial Needle Biopsy .................................. 93
3.3 Endobronchial Ultrasound......................................... 94
3.4 Esophageal Endoscopic Ultrasound .......................... 94 Abbreviations
3.5 Cervical Mediastinoscopy ......................................... 94
3.6 Mediastinotomy ......................................................... 95 AJCC American Joint Committee on Cancer
3.7 Extended Cervical Mediastinoscopy......................... 95 CT Computed tomography
3.8 Video-Assisted Thoracic Surgery ............................. 95 EBUS Endobronchial ultrasound
3.9 Limitations of the Literature on Individual EUS Esophageal endoscopic ultrasound
Mediastinal Staging Modalities................................. 95
IASLC International Association for the Study of
4 Randomized Trials of Staging Algorithms ........... 96 Lung Cancer
5 Staging in the Community at Large ..................... 97 UICC International Union Against Cancer
6 General Recommendations ..................................... 97
MRI Magnetic resonance imaging
NSCLC Non-small cell lung cancer
References.......................................................................... 97
PET Positron emission tomography
SCLC Small cell lung cancer
TBNBx Transbronchial needle biopsy
TTNBx Transthoracic needle biopsy
VATS Video-assisted thoracic surgery

1 Introduction
F. Farjah  V. W. Rusch (&)
Thoracic Surgery Service, The purpose of lung cancer staging is to aid thera-
Memorial Sloan-Kettering Cancer Center, peutic decision-making and provide the patient with a
1275 York Avenue, New York,
NY 10065, USA prognosis. ‘‘Surgical staging’’ often refers to the
e-mail: ruschv@mskcc.org combined information obtained from the clinical

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 89
DOI: 10.1007/174_2011_261, Ó Springer-Verlag Berlin Heidelberg 2011
90 F. Farjah and V. W. Rusch

findings at operation and from the pathologic review Methods for determining T-stage include com-
of a surgically removed specimen. For the purposes of puted tomography (CT), magnetic resonance imaging
this chapter, ‘‘surgical staging’’ refers to any invasive (MRI), and surgical exploration and/or resection.
attempt to obtain tissue for pathologic review. Cangemi et al. (1996) reviewed the literature and their
Surgical staging is only one component of a multi- own experience and found that the sensitivity of CT
modality approach to accurate staging. Accuracy has for detection of T3 or T4 disease was highly variable
a direct bearing on appropriate treatment allocation. and generally low—38–97% for T3 and 31–78% for
Whereas understaged patients may unnecessarily be T4. MRI had a sensitivity of 84% for correct
exposed to the risks of surgical intervention without T-staging in one series (Manfredi et al. 1996). The
benefit, overstaged patients may be deprived a development of higher quality scanners over time has
potentially curative resection. The goal of this chapter potentially improved the accuracy of CT and MRI.
is to (1) review the state-of-the-art in invasive lung Given the inaccuracy of radiographic T-staging,
cancer staging, (2) critically examine the evidence invasive methods are often needed to resolve uncer-
supporting the use of individual staging modalities tainty over T-status. Endoscopic (esophageal) ultra-
and their use in the context of multi-modality staging sound (EUS) has been evaluated as a minimally
algorithms. invasive option for T-staging, though it does not
appear to be superior to imaging given a sensitivity of
88% (Varadarajulu et al. 2004). Surgical exploration
2 Stage Classification is the standard for determining gross invasion. Among
and Determination patients with radiographically suspected T4 disease,
29–50% did not have T4 disease on exploration
The 7th editions of the staging manuals of the (Sebastián-Quetglás et al. 2003; Eggeling et al. 2002;
American Joint Committee on Cancer (AJCC) and De Giacomo et al. 1997). A video-assisted thoraco-
International Union Against Cancer (UICC) use scopic (VATS) approach to exploration is less morbid
information about the primary site (T), regional nodes than a standard thoracotomy, but it is unknown
(N), and distant metastases (M) to establish the stage whether the ability to assess T-status is equivalent
of lung cancer (American Joint Committee on Cancer among these two approaches. Resection is the gold
2010; International Union Against Cancer 2009). standard for T-staging as it allows for pathologic
Unlike prior editions based on single institution data, assessment of size, microscopic invasion, and/or
the 7th edition classifications were based on an visceral pleural involvement.
analysis of the International Association for the Study
of Lung Cancer (IASLC) database. Included in that
dataset were 81,015 lung cancer patients from 46 2.2 N-Stage
institutions and 19 countries. The analysis sought to
identify subgroups based on TNM categories pre- N-stage is determined by involvement of nodes
dicting a unique prognosis (survival rate). spanning an area from the supraclavicular region to
the diaphragm. The IASLC recently resolved
differences between two widely used lymph node
2.1 T-Stage classification schemes—the Naruke map and the
Mountain–Dresler modification of the American
Determinants of T-stage include tumor size, location, Thoracic Society map (American Joint Committee on
sequelae of airway obstruction (atelectasis, pneumo- Cancer 2010). Both the UICC and the AJCC now
nitis, etc.), and/or invasion into adjacent structures. recommend the use of the IASLC map for describing
Location is defined by involvement of the main stem nodal involvement in lung cancer (Fig. 1).
bronchus and distance from the carina. Invasion can Palpable cervical or supraclavicular lymph nodes
involve virtually any structure in the chest, including may be sampled via fine needle aspiration, core nee-
the pleura, great vessels, heart, esophagus, diaphragm, dle biopsy, or incisional/excisional surgical biopsy in
nerves (i.e., phrenic, recurrent laryngeal, etc.), chest the outpatient setting. The latter two approaches yield
wall (i.e., ribs, intercostal muscles, etc.), and vertebrae. more tissue and allow for an architectural and
Surgical Staging of Lung Cancer for Advances in Radiation Oncology 91

Fig. 1 The IASLC/MSKCC lymph node map. Reprinted with permission courtesy of the international Association for the study of
Lung Cancer. Copyright Ó 2009 Memorial Sloan-Kettering Cancer Center
92 F. Farjah and V. W. Rusch

molecular analysis of the material. A positive result resources, such as a cytotechnologist who can be
indicates N3 (stage IIIB) disease precluding the need present to evaluate specimen quality during the pro-
for further nodal staging. cedure. As a result, not all staging procedures are
Non-invasive and invasive staging modalities are accessible to all providers and patients. Finally, all
available to evaluate mediastinal nodes. A key invasive staging carries a small risk of complications.
difference between the two is that mediastinal staging Like T-staging, the decision to pursue invasive stag-
allows for tissue diagnosis prior to instituting treat- ing of the mediastinum ultimately hinges on whether
ment. Two meta-analyses revealed that CT has a the information gained will change management.
sensitivity of 51–61% and specificity of 79–85% for The role of intraoperative nodal staging among
detecting mediastinal lymph node metastases, whereas those eligible for curative resection is rapidly evolv-
positron emission tomography (PET) has a sensitivity ing. Results of a randomized trial of mediastinal
of 74–85% and specificity of 85–90% (Silvestri et al. lymph node sampling versus dissection revealed no
2007; Gould et al. 2003). Accordingly, up to 26 and differences in morbidity or mortality (Allen et al.
10% of patients may be understaged and overstaged, 2006) and no differences in survival in patients with
respectively. Most reviews and practice guidelines N0-1 (non-hilar) lymph disease even though lymph
recommend invasive staging for patients with abnor- node dissection identified additional metastatic nodes
mal nodes on PET (Silvestri et al. 2007; Gould et al. in a small proportion of patients (Darling et al. 2010).
2003; Ettinger et al. 2010). The significance of un- A retrospective review of national registry data
derstaging is controversial because it is unclear to revealed an association between an increasing number
what degree induction therapy as opposed to adjuvant of lymph nodes and higher survival rates (Ludwig
therapy affects long-term outcomes (Gilligan et al. et al. 2005). For the mean time, the general recom-
2007; Song et al. 2010; Felip et al. 2010). Neverthe- mendation from the AJCC is to remove at least 6
less, since practice guidelines recommend neoadju- lymph nodes and to sample or dissect lymph nodes
vant therapy for patients with stage IIIA lung cancer from 2R, 4R, 7, 10R, and 11R for right-sided tumors,
(Ettinger et al. 2010; Robinson et al. 2007), it is and from 5, 6, 7, 10L, and 11L for left-sided tumors
desirable to avoid understaging. Approximately (American Joint Committee on Cancer 2010).
6–16% of patients without evidence of N2/N3 disease Regardless of side, lower lobe tumors should also
on PET will have evidence of occult N2 disease at the include evaluation of the ipsilateral level 9 node.
time of resection (Al-Sarraf et al. 2008; Cerfolio et al.
2006; Defranchi et al. 2009; Kanzaki et al. 2011; Lee
et al. 2007). Risk factors for occult N2 include location 2.3 M-Stage
of the primary (central versus peripheral), upper lobe
primaries, larger tumors, adenocarcinoma, high stan- M-stage is determined by extra-thoracic spread of
dardized uptake values of the primary on PET, PET disease, for instance to the brain, bones, adrenal
positive N1 nodes, or enlarged nodes on CT (Al-Sarraf glands, contralateral lung, liver, pericardium, kidneys,
et al. 2008; Cerfolio et al. 2006; Defranchi et al. 2009; and/or subcutaneous tissues, or pleural and/or peri-
Kanzaki et al. 2011; Lee et al. 2007). In patients with cardial effusions. If a tissue diagnosis already exists
one or more of these clinical features, invasive medi- based on the primary tumor or nodal disease, and
astinal staging should be considered despite normal there is a high suspicion for metastatic disease based
mediastinal lymph nodes on PET. on clinical and radiographic criteria, then it is not
Invasive mediastinal staging definitively addresses necessary to establish M-stage on pathologic grounds.
the mediastinum with several important caveats. No However, when faced with uncertainty about the
single staging modality can access all nodal stations presence of metastatic disease, the basis for obtaining
(Table 1). Thus, no one invasive staging modality can tissue confirmation is choosing the site that offers the
be used to evaluate all lung cancer patients, and in highest probability of tissue yield with the lowest risk
some cases more than one invasive staging modality to the patient.
may be required. Some invasive staging procedures— Examples of procedures used to establish meta-
such as EUS and endobronchial bronchial ultrasound static disease include a CT-guided biopsy, excisional/
(EBUS)—require technical expertise and unique incisional soft tissue biopsy, thoracentesis, VATS,
Surgical Staging of Lung Cancer for Advances in Radiation Oncology 93

Table 1 Mediastinal lymph nodes accessible by individual staging modalities


2R 2L 4R 4L 5 6 7 8 9R 9L
Transthoracic needle biopsy X X X X
Transbronchial needle biopsy X X X
Endobronchial ultrasound guided biopsy X X X
Esophageal endoscopic ultrasound guided biopsy X X X X X X X
Cervical mediastinoscopy X X X X X
Mediastinotomy X X
Extended cervical mediastinoscopy X X X X X X X
Video-assisted thoracoscopic node biopsy (right) X X X X
Video-assisted thoracoscopic node biopsy (left) X X X X X X

pericardiocentesis, or pericardial window. CT-guided


biopsy is commonly used as an outpatient procedure 3 Invasive Mediastinal Staging
for obtaining tissue from viscera, bone, and deep soft Modalities
tissues. For more superficial lesions, an incisional/
excisional biopsy may be performed in the clinic. 3.1 Transthoracic Needle Biopsy
EUS uses ultrasound visualization to direct a needle
across the esophagus and/or stomach to access Transthoracic Needle Biopsy (TTNBx) is an outpatient
radiographic abnormalities in the retroperitoneum, procedure using CT guidance to direct a small needle
left lobe of the liver, and/or left adrenal gland into enlarged mediastinal lymph nodes. In a review of 5
(LeBlanc et al. 2003). Thoracentesis is an office studies totaling 215 patients, all with clinical N2/N3
procedure involving the introduction of a soft, disease, the sensitivity and specificity of TTNBx were
removable catheter into the pleural space for evacu- 89 and 100%, respectively (Detterbeck et al. 2007). The
ation of an effusion. A patient may require a thora- prevalence of mediastinal nodal disease was 81%, and
centesis for a symptomatic or newly diagnosed only 75% of patients had lung cancer with up to half of
effusion. Thoracentesis can establish a diagnosis of those patients having small cell lung cancer (SCLC).
M1 disease in 80% of patients who truly have Approximately 10% of patients required an additional
malignant pleural effusions, though it may require procedure to decompress a pneumothorax.
three separate pleural fluid specimens reviewed by an
experienced cytologist (Rivera et al. 2007). Per-
forming a VATS procedure for effusions has the 3.2 Transbronchial Needle Biopsy
added benefit of inspecting the pleural, mediastinal,
and diaphragmatic surfaces for disease. In patients Transbronchial Needle Biopsy (TBNBx) uses a flexible
with cytologically negative effusions, VATS discov- bronchoscope to pass an aspirating needle across the
ered pleural disease in 60% of patients (De Giacomo airway into a subcarinal (or sometimes a paratracheal)
et al. 1997). Pericardiocentesis is an outpatient node for the purposes of sampling. Though a blind
procedure involving insertion of a soft, removable procedure, pre-procedure CT imaging informs target-
catheter into the pericardial space for evacuation of an ing. This outpatient procedure is performed in either an
effusion. A pericardial window is a surgically per- endoscopy suite or operating room. Challenges asso-
formed procedure where a portion of the pericardium ciated with sampling paratracheal nodes include
is removed in addition to evacuation of fluid. This knowing precisely where the nodes are with broncho-
procedure may be performed either through a subxi- scopic visualization only and the angulation required to
phoid incision or a right or left VATS procedure. access such nodes. Risks include major hemorrhage,
Pericardial procedures are most often performed to pneumothorax, and pneumomediastinum, with an
treat a symptomatic effusion or cardiac tamponade overall major complication rate of 0.3% (Holty et al.
rather than to establish the presence of M1 disease. 2005).
94 F. Farjah and V. W. Rusch

Two systematic reviews provide information about (360 degree cross-sectional view) or linear (180
the diagnostic performance of TBNBx. The first iden- degree capital view) ultrasound probe with pulse
tified a total of 13 studies stratified into Tier 1 and 2 wave and color Doppler capabilities. This procedure
investigations based on the quality of the study (Holty is performed on an outpatient basis. Under direct
et al. 2005). Tier 1 studies yielded a sensitivity and ultrasound visualization a needle is passed across the
specificity of 36 and 98%, respectively, whereas in Tier esophagus to access nodes in the mediastinum
2 investigations the sensitivity was 82% (specificity not (LeBlanc et al. 2003). Complications associated with
calculated because the lack of surgical confirmation in EUS have been reported as rare (0.8%) and minor
Tier 2 studies). The prevalence of nodes were markedly (fever, strider, sore throat, and cough) (Micames et al.
different between the 2 tiers (36 and 81%), suggesting 2007).
significant differences in the underlying study popula- Two systematic reviews provide information about
tions. Detterbeck et al. (2007) reviewed 17 papers the utility of EUS. Micames et al. (2007) report on 18
including 1,339 patients and calculated a sensitivity studies with 1201 patients using both radial and linear
and specificity of 78 and 100%, respectively. probes revealing a sensitivity and specificity of 83 and
97%, respectively. Sensitivity and specificity varied
with the presence or absence of enlarged nodes on
3.3 Endobronchial Ultrasound CT. Detterbeck et al. (2007) examined 16 studies with
973 patients and found a similar sensitivity and
Endobronchial Ultrasound (EBUS) involves a special specificity of 84 and 99.5%, respectively. The prev-
bronchoscope—similar in size to a standard adult alence of positive mediastinal nodes in that review
bronchoscope—with a curvilinear probe that provides a was 61%, and the authors again observed variation in
50 degree linear continuous B-mode ultrasound image performance characteristics based on radiographic
with color Doppler capability. There is a biopsy channel appearance of nodes.
that allows passage of a 22-gauge biopsy needle at a 30
degree angle to the long axis of the bronchoscope.
Optionally, a latex balloon can be placed over the end of 3.5 Cervical Mediastinoscopy
the ultrasound probe to facilitate a fluid interface
between the probe and tracheal or bronchial wall. EBUS Mediastinoscopy is a surgical procedure that allows
is an outpatient procedure performed in either the for direct visualization and biopsy of paratracheal and
operating room or endoscopy suite. Subcarinal and level subcarinal nodes. The patient is administered a gen-
10 or 11 nodes are relatively straightforward to access, eral anesthetic, a 2 cm transverse cervical incision is
whereas sampling paratracheal nodes may be difficult made, and a mediastinoscope is passed just anterior to
because of the angle at which the scope must be placed to the trachea into the mediastinum. Mediastinoscopy is
visualize and access these nodes. an outpatient procedure (Cybulsky and Bennett 1994;
Two systematic reviews provide information on Vallières et al. 1991), although it can also be per-
the performance characteristics of this staging formed immediately prior to resection. Risks associ-
modality. The first review summarized 8 studies with ated with mediastinoscopy among a series of 2,145
918 patients and found a sensitivity and specificity of cases included bleeding (0.33%), vocal cord dys-
90 and 100%, respectively. Notably, 14–30% of function (0.55%), tracheal injury (0.09%), pneumo-
patients had SCLC and the prevalence of nodes in this thorax (0.09%), and death (0.05%) (Lemaire et al.
study was 68%. The second review summarized 10 2006).
studies and found sensitivity and specificity to be 88 Detterbeck et al. (2007) reviewed 19 studies
and 100%, respectively (Adams et al. 2009). including 6505 patients and determined a sensitivity
of 78%. The prevalence of mediastinal node
involvement was 39%. With the addition of a video-
3.4 Esophageal Endoscopic Ultrasound endoscope, the sensitivity was 90%. A national study
revealed that just under half of patients undergoing
Esophageal Endoscopic Ultrasound (EUS) is per- mediastinoscopy had documented evidence of lymph
formed using a special endoscope with either a radial node material submitted to pathology. The proportion
Surgical Staging of Lung Cancer for Advances in Radiation Oncology 95

of patients with submitted lymph node material was through small incisions and with the aid of a
higher when performed at teaching centers and video-thoracoscope. This procedure requires general
comprehensive community cancer centers suggesting anesthesia and single lung ventilation. Patients
that the diagnostic yield of mediastinoscopy is oper- typically stay in hospital for one day, though it can be
ator dependent (Little et al. 2005). performed as an outpatient procedure. A right-sided
VATS allows access to levels R4, 7, R9, and R10,
whereas a left-sided VATS provides access to levels
3.6 Mediastinotomy 5, 6, 7, L8, L9, and L10. Complications occurred in
less than 2% of patients (Detterbeck et al. 2007).
Mediastinotomy is a surgical procedure that allows Detterbeck et al. (2007) summarized the results of
direct visualization and biopsy of mediastinal nodes in 7 studies including 419 patients undergoing VATS for
the aortopulmonary window (levels 5, 6). This pro- mediastinal lymph node staging. The sensitivity and
cedure is particularly useful for sampling nodes specificity were 75 and 100%, respectively (Detter-
associated with a left upper lobe primary as these beck et al. 2007). The authors note inexplicable and
tumors have a predilection to drain into nodes in the wide variation in the sensitivity of VATS across
aortopulmonary window. After administration of studies (37–100%), which likely reflects differences
general anesthesia, a 3–6 cm transverse incision is in surgeon experience and patient selection.
made over the second intercostal space and a medi-
astinoscope is passed into the mediastinum. The sec-
ond costal cartilage may be resected to improve
exposure. Risks associated with this procedure include 3.9 Limitations of the Literature
bleeding, infection, injury to the phrenic nerve, and on Individual Mediastinal Staging
pneumothorax. A systematic review found the sensi- Modalities
tivity of mediastinotomy to be 87% (Detterbeck et al.
2007). It should be noted that the prevalence of nodes There are very specific criteria for cohort selection in
based on imaging ranged from 47 to 77% and that most studies of individual mediastinal staging
approximately one-fifth of patients had SCLC. modalities. Most studies evaluate operated patients
because surgical nodal assessment is considered the
‘‘gold standard’’. By virtue of being selected for
3.7 Extended Cervical Mediastinoscopy
surgical therapy, most of these patients had a low pre-
treatment likelihood of advanced or late stage disease.
Extended cervical mediastinoscopy is an alternative
Furthermore, operated patients only account for
surgical approach to visualize and sample nodes in the
approximately 25% of all non-small cell lung cancer
aortopulmonary window. The procedure is similar to
(NSCLC) patients (Little et al. 2007; Farjah et al.
standard cervical mediastinoscopy (see above) except
2009). Evaluating performance characteristics among
that the mediastinoscope is directed laterally to the
operated patients makes it difficult to generalize the
aortic arch. This procedure is now rarely performed
results to all lung cancer patients. Another criterion for
and has been largely supplanted by EBUS, EUS and
cohort selection is including only those patients with
VATS (video-assisted thoracic surgery). The sensi-
radiographically abnormal nodes in nodal stations
tivity ranges from 45 to 51% given a prevalence
accessible by the staging modality of interest. For
ranging from 29 to 48% (Detterbeck et al. 2007).
instance, studies of TTNBx predominantly included
Though complications were only reported in 0.3% of
patients with bulky N2/N3 disease (Detterbeck et al.
patients, the events were severe involving one aortic
2007). Though this approach is rationale for the study
injury and one stroke (Detterbeck et al. 2007).
of diagnostic accuracy, it is difficult to compare per-
formance characteristics of individual staging modal-
3.8 Video-Assisted Thoracic Surgery ities across studies. There are also significant
differences in patient characteristics across investiga-
Video-assisted thoracic surgery (VATS) is a surgical tions that make it difficult to compare results across
procedure involving access to the hemithorax studies, particularly with regard to the proportion of
96 F. Farjah and V. W. Rusch

NSCLC and SCLC patients and prevalence of medi- Fischer et al. 2009). It also remains uncertain whether
astinal nodes (Detterbeck et al. 2007). PET decreases the overall number of staging tests
Another problem with the existing body of litera- performed (Herder et al. 2006; Maziak et al. 2009).
ture is the lack of a uniform reference standard. PET incorrectly upstages patients more frequently
Surgery is often times regarded as the ‘‘gold stan- than conventional work-up with CT, bone scans, and
dard’’ for assessing the performance of a staging selective mediastinoscopy (Maziak et al. 2009),
modality, but most studies do not detail the conduct potentially requiring more diagnostic investigations to
of an operation with regard to the extent of intra- confirm PET results. There is no evidence that PET
operative lymph node evaluation (stations examined, affects survival (Viney et al. 2004) or direct medical
dissections versus sampling, etc.). The extent of costs (Herder et al. 2006), though these endpoints
lymph node evaluation likely varies significantly were secondary outcomes in the trials that evaluated
across surgeons and institutions. Recently emerging them. One study demonstrated that PET leads to a
data may define a future standard for intraoperative clinically important but statistically non-significant
nodal assessment (Darling et al. 2010). Without a lower median survival (Fischer et al. 2009).
uniform reference standard, it is difficult to compare Several trials show that the addition of endoscopic
the performance characteristics of individual staging guided modalities to ‘‘conventional’’ staging led to
modalities across studies. higher sensitivity for N2/N3 detection and fewer
Staging modalities are often considered not only in futile thoracotomies and surgical interventions
terms of their diagnostic accuracy but also their risks. (Annema et al. 2010; Tournoy et al. 2008; Larsen
Complications are not reported in a uniform fashion, et al. 2005). When compared to mediastinoscopy
and in some cases they are not reported at all. Criteria among patients with CT and/or PET evidence of
exist for grading the severity of adverse events mediastinal nodal disease, combination EUS/EBUS
following procedures and could be used explicitly in resulted in greater sensitivity for N2/N3 disease and
investigations of invasive staging modalities (CTCAE fewer unnecessary thoracotomies without significant
and National Cancer Institute 2006). Another impor- differences in the median number of nodal stations
tant aspect of measuring risk is establishing a set sampled or complications (Annema et al. 2010).
period of time for follow-up, particularly since most A small study compared EUS versus mediastinoscopy
staging procedures occur on an outpatient basis. In in a similar cohort of lung cancer patients and found
order to fairly weigh the risks and benefits of com- that the number of surgical procedures was lower in
peting invasive staging procedures, a high level of the EUS group (Tournoy et al. 2008). However, the
rigor must be applied to ascertaining complications. number of mediastinal nodes sampled was signifi-
cantly higher in the mediastinoscopy group. Pre-
liminary results from a different trial found that
4 Randomized Trials of Staging incorporating routine EUS into a conventional algo-
Algorithms rithm consisting of CT and selective TTNBx,
TBNBx, and mediastinoscopy led to fewer unneces-
To date, there are eight randomized trials comparing sary thoracotomies. Only one-third of patients
different lung cancer staging algorithms. Some studies underwent PET because they were double enrolled in
evaluated the use of PET. These trials are also another trial, and the frequency of PET was higher in
reviewed because invasive staging was a component the routine EUS group.
of the overall staging strategy. Despite a randomized study design, there are a
Most, though not all trials, suggest that incorpo- number of important limitations that limit the pooling
rating PET into a staging algorithm reduces the of results and ability to generalize the findings.
number of futile or unnecessary thoracotomies Inclusion criteria varied across investigations. The
(van Tinteren et al. 2002; Herder et al. 2006; Maziak control arms of the trials often refer to a ‘‘conven-
et al. 2009; Fischer et al. 2009). There is conflicting tional’’ or ‘‘standard’’ staging algorithm based on
data on whether PET does so through a mechanism of local or national guidelines. Yet, the control algo-
better detection of N2/N3 disease (Herder et al. 2006; rithms varied significantly across studies suggesting
Surgical Staging of Lung Cancer for Advances in Radiation Oncology 97

there is no standard, or at least none across countries


and/or the decade of study. Primary endpoints also 6 General Recommendations
varied significantly across studies including for
instance diagnostic performance, futile thoracotomies The UICC and AJCC 7th edition staging manuals
(variably defined), and number of staging modalities define the current standard for classifying lung cancer
utilized to arrive at a final clinical stage. Finally, it is stage (American Joint Committee on Cancer 2010;
unclear whether the community at large will have International Union Against Cancer 2009). All
access to the unique resources and expertise necessary patients should undergo non-invasive staging with CT
to utilize certain staging modalities, such as EUS and and PET. Those with clear evidence of widely met-
EBUS. astatic disease should have tissue confirmation of a
cancer diagnosis and a referral to medical and radia-
tion oncology. For cases where there is a plausible
5 Staging in the Community at Large alternative explanation for what appears to be meta-
static disease on CT/PET, invasive staging is indi-
Few studies describe how patients in the community cated to rule out the possibility of metastatic cancer.
at large are actually staged. Little et al. (2007) used In an otherwise resectable patient with PET evidence
the National Cancer Database to describe the use of of abnormal nodes, invasive staging is indicated to
individual staging modalities among a nationally rule in or out the possibility of mediastinal nodal
representative sample of lung cancer patients cared metastases. Invasive mediastinal staging should be
for at 729 hospitals in 2001. Among 40,909 patients, considered even if PET reveals no uptake in medi-
93 and 19% underwent CT and PET, respectively. CT astinal nodes when the primary tumor is an adeno-
and PET revealed abnormal mediastinal lymph nodes carcinoma, large, centrally located or in the upper
in 56 and 51% of patients, respectively. Overall, 20% lobes, and/or has a high standardized uptake value on
of patients underwent mediastinoscopy. Using the PET, and/or there is PET evidence of N1 disease or
same database but evaluating only 11,688 resected CT evidence of abnormal mediastinal nodes. Medi-
patients, 92% underwent CT and 26% underwent PET astinoscopy, EUS, and/or EBUS are all legitimate
(Little et al. 2005). Abnormal mediastinal nodes were options for evaluating mediastinal lymph nodes. The
detected in 27 and 28% of patients by CT and PET, decision to use one or more such modalities should be
respectively. Overall, 27% of patients underwent guided by the nodal distribution of suspected disease
mediastinoscopy, but only 47% of these patients had and the availability of appropriate resources and
documented lymph node material submitted for expertise to safely and effectively conduct these
pathologic review. procedures. For patients with radiographic evidence
Farjah et al. (2009) evaluated multi-modality of invasion, who are otherwise resectable and have
mediastinal staging in 43,912 elderly NSCLC patients had an extensive evaluation ruling out mediastinal
between 1998 and 2002 using the Surveillance, Epi- node involvement, exploration is indicated to deter-
demiology, and End-Results database linked to mine resectability.
Medicare claims (Farjah et al. 2009). CT, PET, and
invasive staging—defined by mediastinoscopy,
mediastinotomy, VATS, or EUS—were evaluated.
Most patients (77%) underwent staging with CT only, References
whereas only 21 and 2% underwent bi- and tri-
modality staging, respectively. Over the 5 year study Adams K, Shah PL, Edmonds L, Lim E (2009) Test perfor-
mance of endobronchial ultrasound and transbronchial
period, there was a dramatic decrease in the use of needle aspiration biopsy for mediastinal staging in patients
single modality staging from 90 to 67% coincident with lung cancer: systematic review and meta-analysis.
with a rise in bimodality staging from 10 to 30%. The Thorax 64:757–762
rise in bimodality staging was accounted for by a Allen MS, Darling GE, Pechet TTV, Mitchell JD, Herndon JE
II, Landreneau RJ, Inculet RI, Jones DR, Meyers BF,
rapid increase in the use of PET from 2 to 30%. Over Harpole DH, Putnam JB Jr, Rusch VW, The ACOSOG
the same period of time, the use of invasive staging Z0030 Study Group (2006) Morbidity and mortality of
did not change (9–8%). major pulmonary resections in patients with early-stage
98 F. Farjah and V. W. Rusch

lung cancer: initial results of the randomized, prospective Farjah F, Flum DR, Ramsey SD, Heagerty PJ, Symons RG,
ACOSOG Z0030 trial. Ann Thorac Surg 81:1013–1020 Wood DE (2009) Multi-modality mediastinal staging for
Al-Sarraf N, Aziz R, Gately K, Lucey J, Wilson L, McGovern E, lung cancer among medicare beneficiaries. J Thorac Oncol
Young V (2008) Pattern and predictors of occult mediastinal 4(3):355–363
lymph node involvement in non-small cell lung cancer Felip E, Rosell R, Maestre JA, Rodriguez-Paniagua JM, Morán
patients with negative mediastinal uptake on positron emis- T, Astudillo J, Alonso G, Borro JM, González-Larriba JL,
sion tomography. Eur J Cardiothorac Surg 33(1):104–109 Torres A, Camps C, Guijarro R, Isla D, Aguiló R, Alberola V,
American Joint Committee on Cancer (2010) AJCC Cancer Sánchez-Palencia A, Sánchez JJ, Hermosilla E, Massuti B,
Staging Manual, 7th edn. Springer, New York The Spanish Lung Cancer Group (2010) Preoperative
Annema JT, Van Meerbeeck JP, Rintoul RC, Dooms C, chemotherapy plus surgery versus surgery plus adjuvant
Deschepper E, Dekkers OM, De Leyn P, Braun J, Carroll NR, chemotherapy versus surgery alone in early-stage
Praet M, de Ryck F, Vansteenkiste J, Vermassen F, Versteegh non-small cell lung cancer. J Clin Oncol 28(19):3138–
MI, Veseliç M, Nicholson AG, Rabe KF, Tournoy KG (2010) 3145
Mediastinoscopy vs endosonography for mediastinal nodal Fischer B, Lassen U, Mortensen J, Larsen S, Loft A, Bertelsen
staging of lung cancer: a randomized trial. J Am Med Assoc A, Ravn J, Clementsen P, Hogholm A, Larsen K, Rasmus-
304(20):2245–2252 sen T, Keiding S, Dirksen A, Gerke O, Skov B, Steffensen I,
Cangemi V, Volpino P, Drudi FM, D’Andrea N, Cangemi R, Hansen H, Vilmann P, Jacobsen G, Backer V, Maltbaek N,
Piat G (1996) Assessment of the accuracy of diagnostic Pedersen J, Madsen H, Nielsen H, Hojgaard L (2009)
chest CT scanning. impact on lung cancer management. Int Preoperative staging of lung cancer with combined PET-
Surg 81(1):77–82 CT. N Engl J Med 361:32–39
Cerfolio RJ, Bryant AS, Eloubeidi MA (2006) Routine medias- Gilligan D, Nicolson M, Smith I, Groen H, Dalesio O,
tinoscopy and esophageal ultrasound fine-needle aspiration in Goldstraw P, Hatton M, Hopwood P, Manegold C, Schra-
patients with non-small cell lung cancer who are clinically N2 mel F, Smit H, van Meerbeeck J, Nankivell M, Parmar M,
negative. a prospective study. Chest 130:1791–1795 Pugh C, Stephens R (2007) Preoperative chemotherapy in
CTCAE and National Cancer Institute (2006) Common termi- patients with resectable non-small cell lung cancer: results
nology criteria for adverse events v.3.0. http://ctep.cancer. of the MRC LU22/NVALT 2/EORTC 08012 multicentre
gov/forms/CTCAEv3.pdf. Accessed 2006 randomised trial and update of systematic review. Lancet
Cybulsky IJ, Bennett WF (1994) Mediastinoscopy as a routine 369:1929–1937
outpatient procedure. Ann Thorac Surg 58:176–178 Gould MK, Kuschner WG, Rydzak CE, Maclean CC, Demas AN,
Darling GE, Allen MS, Decker PA, Ballman K, Malthaner RA, Shigemitsu H, Chan JK, Owens DK (2003) Test perfor-
Inculet RI, Jones DR, McKenna RJ, Landreneau RJ, Putnam mance of positron emission tomography and computed
JB Jr (2010) Number of lymph nodes harvested from a tomography for mediastinal staging in patients with non-
mediastinal lymphadenectomy: results of the randomized, small cell lung cancer. a meta-analysis. Ann Int Med
prospective ACOSOG Z0030 trial. doi: 10.1378/chest. 139:879–892
10-0859 Herder GJM, Kramer H, Hoekstra OS, Smit EF, Pruim J,
De Giacomo T, Rendina EA, Venuta F, Della Rocca G, Ricci C van Tinteren H, Comans EF, Verboom P, Uyl-de-Groot CA,
(1997) Thoracoscopic staging of IIIB non-small cell lung Welling A, Paul MA, Boers M, Postmus PE, Teule GJ,
cancer before neoadjuvant therapy. Ann Thorac Surg Groen HJM (2006) Traditional versus up-front [18F] fluo-
64:1409–1411 rodeoxyglucose-positron emission tomography staging of
Defranchi SA, Cassivi SD, Nichols FC, Allen MS, Shen KR, non-small cell lung cancer: a Dutch cooperative randomized
Deschamps C, Wigle DA (2009) N2 disease in T1 non- study. J Clin Oncol 24:1800–1806
small cell lung cancer. Ann Thorac Surg 88(3):924–928 Holty JE, Kuschner WG, Gould MK (2005) Accuracy of
Detterbeck FC, Jantz MA, Wallace M, Vansteenkiste J, transbronchial needle aspiration for mediastinal staging of
Silvestri GA, The American College of Chest Physicians non-small cell lung cancer: a meta-analysis. Thorax
(2007) Invasive mediastinal staging of lung cancer: ACCP 60(11):949–955
evidence-based clinical practice guidelines (2nd edition). International Union Against Cancer (2009) TNM classification
Chest 132(3 Suppl):202S–220S of malignant tumours, 7th edn. Wiley-Blackwell, Oxford
Eggeling S, Martin T, Böttger J, Beinert T, Gellert K (2002) Kanzaki R, Higashiyama M, Fujiwara A, Tokunaga T, Maeda J,
Invasive staging of non-small cell lung cancer—a prospec- Okami J, Kozuka T, Hosoki T, Hasegawa Y, Takami M,
tive study. Eur J Cardiothorac Surg 22(5):679–684 Tomita Y, Kodama K (2011) Occult mediastinal lymph
Ettinger DS, Akerley W, Bepler G, Blum MG, Chang A, node metastasis in NSCLC patients diagnosed as clinical
Cheney RT, Chirieac LR, D’Amico TA, Demmy TL, Ganti N0–1 by preoperative integrated FDG-PET/CT and CT: risk
AK, Govindan R, Grannis FW Jr, Jahan T, Jahnanzeb M, factors, pattern, and histopathological study. Lung Cancer
Johnson DH, Kessinger A, Komaki R, Kong FM, Kris MG, 71(3):333–337
Krug LM, Le Q-T, Lennes IT, Martins R, O’Malley J, Larsen SS, Vilmann P, Krasnik M, Dirksen A, Clementsen P,
Osarogiagbon RU, Otterson GA, Patel JD, Pisters KM, Maltbaek N, Lassen U, Skov BG, Jacobsen GK (2005)
Reckamp K, Riely GJ, Rohren E, Simon GR, Swanson SJ, Endoscopic ultrasound guided biopsy performed routinely
Wood DE, Yang SC, NCCN Non-Small Cell Lung Cancer in lung cancer staging spares futile thoracotomies: prelmi-
Panel Members (2010) Non-small cell lung cancer. J Natl nary results form a randomised clinical trial. Lung Cancer
Compr Canc Netw 8(7):740–801 49(3):377–385
Surgical Staging of Lung Cancer for Advances in Radiation Oncology 99

LeBlanc JK, Espada R, Ergun G (2003) Non-small cell lung based clinical practice guidelines (2nd edition). Chest
cancer staging techniques and endoscopic ultrasound: tissue 132(Suppl 3):243S–265S
is still the issue. Chest 123:1718–1725 Sebastián-Quetglás F, Molins L, Baldó X, Buitrago J, Vidal G,
Lee PC, Port JL, Korst RJ, Liss Y, Meherally DN, Altorki NK The Spanish Video-Assisted Thoracic Surgery Group
(2007) Risk factors for occult mediastinal metastases in (2003) Clinical value of video-assisted thoracoscopy for
clinical stage I non-small cell lung cancer. Ann Thorac Surg preoperative staging of non-small cell lung cancer. a
84:177–181 prospective study of 105 patients. Lung Cancer 42:297–301
Lemaire A, Nikolic I, Petersen T, Haney JC, Toloza EM, Silvestri GA, Gould MK, Margolis ML, Tanoue LT, McCrory D,
Harpole DH Jr, D’Amico TA, Burfeind WR (2006) Nine- Toloza E, Detterbeck F, The American College of Chest
year single center experience with cervical mediastinos- Physicians (2007) Noninvasive staging of non-small cell lung
copy: complications and false negative rate. Ann Thorac cancer: ACCP evidenced-based clinical practice guidelines
Surg 82(4):1185–1189 (2nd edition). Chest 132(3 Suppl):178S–201S
Little AG, Rusch VW, Bonner JA, Gaspar LE, Green MR, Song WA, Zhou NK, Wang W, Chu XY, Liang CY, Tian XD,
Webb WR, Stewart AK (2005) Patterns of surgical care of Guo JT, Liu X, Liu Y, Dai WM (2010) Survival benefit of
lung cancer patients. Ann Thorac Surg 80:2051–2056 neodjuvant chemotherapy in non-small cell lung cancer: an
Little AG, Gay EG, Gaspar LE, Stewart AK (2007) National updated meta-analysis of 13 randomized control trials.
survey of non-small cell lung cancer in the United States: J Thorac Oncol 5(4):510–516
epidemiology, pathology and patterns of care. Lung Cancer Tournoy KG, de Ryck F, Vanwalleghem LR, Vermassen F,
57(3):253–260 Praet M, Aerts JG, Van Maele G, Van Meerbeeck JP (2008)
Ludwig MS, Goodman M, Miller DL, Johnstone PA (2005) Endoscopic ultrasound reduces surgical mediastinal staging
Postoperative survival and the number of lymph nodes in lung cancer: a randomized trial. Am J Respir Crit Care
sampled during resection of node-negative non-small cell Med 177(5):531–535
lung cancer. Chest 128(3):1545–1550 Vallières E, Pagé A, Verdant A (1991) Ambulatory mediasti-
Manfredi R, Pirronti T, Bonomo L, Marano P (1996) Accuracy noscopy and anterior mediastinotomy. Ann Thorac Surg
of computed tomography and magnetic resonance imaging 52:1122–1126
in staging bronchogenic carcinoma. MAGMA 4(3–4):257– van Tinteren H, Hoekstra OS, Smit EF, van den Bergh HJAM,
262 Schreurs AJM, Stallaert RALM, van Velthoven PCM,
Maziak DE, Darling GE, Inculet RI, Gulenchyn KY, Driedger AA, Comans EFI, Diepenhorst FW, Verboom P, van Mourik JC,
Ung YC, Miller JD, Gu C-S, Cline KJ, Evans WK, Levine MN Postmus PE, Boers M, Teule GJJ, The PLUS Study Group
(2009) Positron emission tomography in staging early lung (2002) Effectiveness of positron emission tomography in the
cancer. Ann Intern Med 151:221–228 preoperative assessment of patients with suspected non-
Micames CG, McCrory DC, Pavey DA, Jowell PS, Gress FG small cell lung cancer: the PLUS multicentre randomised
(2007) Endoscopic ultrasound-guided fine-needle aspiration trial. Lancet 359:1388–1392
for non-small cell lung cancer staging: a systematic review Varadarajulu S, Schmulewitz N, Wildi SF, Roberts S, Ravenel J,
and metaanalysis. Chest 131(2):539–548 Reed CE, Block M, Hoffman BJ, Hawes RH, Wallace MB
Rivera MP, Mehta AC, The American College of Chest (2004) Accuracy of EUS in staging of T4 lung cancer.
Physicians (2007) Initial diagnosis of lung cancer: ACCP Gastrointest Endosc 59:345–348
evidence-based clinical practice guidelines (2nd edition). Viney RC, Boyer MJ, King MT, Kenny PM, Pollicino CA,
Chest 132(Suppl 3):131S–148S McLean JM, McCaughan BC, Fulham MJ (2004) Random-
Robinson LA, Ruckdeschel JC, Wagner H Jr, Stevens CW, The ized controlled trial of the role of positron emission
American College of Chest Physicians (2007) Treatment of tomography in the management of Stage I and II non-small
non-small cell lung cancer - stage IIIA: ACCP evidence- cell lung cancer. J Clin Oncol 22:2357–2362
Lung Cancer Surgery
Sidhu P. Gangadharan, Walter J. Lech, and David J. Sugarbaker

Contents Abstract
Lung cancer is the most common cancer world-
1 Introduction.............................................................. 103 wide. Current estimates project over 222,000 new
2 Preoperative Assessment......................................... 104 cases of lung cancer in the US alone in 2010,
2.1 Diagnosis.................................................................... 104 amounting to over 157,000 deaths. Despite a
2.2 Preoperative Fitness................................................... 108 modest improvement in 5-year survival over the
3 Operative Strategy................................................... 109 past three decades, deaths from lung cancer con-
3.1 Nomenclature and Anatomy ..................................... 109 tinue to exceed cancer deaths from all other sites.
3.2 Extent of Resection ................................................... 109 This chapter updates the role of surgical resection
3.3 Technique of Resection............................................. 110
3.4 Postoperative Course ................................................. 111 in the management of lung cancer. Among the
3.5 Video-Assisted Thoracoscopic Surgery .................... 112 topics reviewed are the diagnostic work-up and
3.6 Radiation Therapy for Patients Undergoing Lung preoperative assessment of suitability for resection,
Resection: T3 Tumors ............................................... 113 open and video-assisted thoracoscopic operative
References.......................................................................... 113 strategies, and recent changes in both the lung
cancer staging and histological classification sys-
tems. Indications for radiation therapy in selected
S. P. Gangadharan
Division of Thoracic Surgery, patients with T3 tumors are also discussed.
Beth Israel Deaconess Medical Center,
185 Pilgrim Road, W/DC 201,
Boston, MA 02115, USA
S. P. Gangadharan
Assistant Professor of Surgery,
1 Introduction
Harvard Medical School,
Boston, MA, USA Lung cancer is the most common cancer in the world,
W. J. Lech  D. J. Sugarbaker (&) with an incidence of 1.52 million cases and
Division of Thoracic Surgery, 1.31 million deaths in the year 2008 (Boyle and Levin
Brigham and Women’s Hospital, 2008). It has been estimated that over 222,000
75 Francis Street, Boston, MA 02115, USA
e-mail: dsugarbaker@partners.org; new cases of lung cancer occurred in the US in
asadams@partners.org 2010, accounting for 15% of all cancer diagnoses
W. J. Lech
(American Cancer Society 2010). More than 157,000
Instructor in Surgery, Americans are expected to die in 2010 as a result,
Harvard Medical School, comprising nearly 28% of all non-melanoma cancer-
Boston, MA, USA related deaths. Over the past three decades the US has
D. J. Sugarbaker experienced a modest, yet statistically significant
The Richard E. Wilson Professor of Surgical Oncology, improvement in 5-year survival, increasing from
Harvard Medical School, Boston, MA, USA

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 103
DOI: 10.1007/174_2011_318,  Springer-Verlag Berlin Heidelberg 2011
104 S. P. Gangadharan et al.

11.5% for patients diagnosed in 1975 to 16.0% in dullness from an effusion, and neck vein distension
2003 (Howlader et al. 2011). However, death from from superior vena cava obstruction. After radiologic
lung cancer still exceeds that from any other site, confirmation of the presence of tumor, a pathological
representing 29% of male and 26% of female cancer diagnosis may be obtained by means of sputum
deaths in 2010 (American Cancer Society 2010). cytology, bronchial washings or brushings, or fine
The World Health Organization has classified the needle aspiration. Bayesian theory has been applied to
histologic subtypes of lung cancer (Travis et al. 2004). the undiagnosed pulmonary nodule to estimate like-
Recently, a multidisciplinary classification of lung lihood of malignancy (Cummings et al. 1986; Gurney
adenocarcinoma was sponsored by the International 1993; Gurney et al. 1993). This approach considers
Association for the Study of Lung Cancer (IASLC), the resulting pre-test probability of malignancy, in
American Thoracic Society, and European Respiratory conjunction with the patient’s operative risk, and
Society (Travis et al. 2011). For resection specimens, stratifies the patient into one of three categories:
the term bronchioloalveolar carcinoma (BAC) has been observation, further non-resectional diagnostic testing
supplanted by adenocarcinoma in situ (AIS) to describe (e.g., sputum cytology, bronchial washings or brush-
small solitary adenocarcinomas with pure lepidic ings, fine needle aspiration, PET scan), or surgical
growth. BAC lesions with less than pure lepidic growth resection (Ost et al. 2003). The patient’s pre-test
are termed either minimally invasive adenocarcinoma probability is highly dependent upon age, smoking
(MIA) or lepidic predominant invasive adenocarci- history, and CT scan characteristics of the lesion
noma based on depth of invasion, with the former being (i.e., size [2 cm, spiculations, and rate of growth).
characterized by a depth B5 mm. Invasive mucinous
adenocarcinoma (formerly mucinous BAC) as its name 2.1.1 Staging
would suggest, is distinguished from MIA by the pres- Once the diagnosis of pulmonary malignancy has
ence of mucin. Other subtypes include colloid, fetal, been made or, conversely, the pre-test probability is
and enteric adenocarcinoma (Travis et al. 2011). sufficient to warrant resection without preoperative
Surgical resection represents the best chance for tissue diagnosis, the patient undergoes a staging
cure of epithelial non-small-cell lung cancers workup to assign prognosis and determine the most
(NSCLC). Unfortunately, over half of patients present appropriate therapy. Verification of clinical stage is
with lesions that are unresectable because of locally central to the development of a treatment plan and
advanced tumor or systemic spread (Howlader et al. may be accomplished using multiple modalities, both
2011; Silvestri et al. 2003). This chapter provides an invasive (e.g., fine needle biopsy, resection) and non-
overview of the important surgical aspects of lung invasive (e.g., CT, MRI, FDG-PET scans). Stages I
cancer therapy, including preoperative assessment, and II NSCLC are treated with surgical resection.
operative strategy, adjuvant and neoadjuvant therapy, Patients with locally advanced Stage III tumors are
and the options for challenging cases such as tumors potential candidates for surgery, depending on the
invading the mediastinum or chest apex. specific aspects of local invasion (e.g., tumor infil-
tration into the chest wall or carina versus involve-
ment of great vessels or heart) or level of nodal
2 Preoperative Assessment metastasis. Stage IV cancers exhibiting extensive
metastatic spread are generally outside the realm of
2.1 Diagnosis the thoracic surgeon, except for palliative measures,
although solitary metastases may not preclude a
Patients presenting with lung cancer are usually potentially curative lung resection. Patients whose
symptomatic and describe a history of cough, weight poor medical condition precludes a pulmonary
loss, or dyspnea in 60–75% of cases (Beckles et al. resection may still benefit from interventions to
2003). Hemoptysis, chest or bone pain, fever, or restore and maintain airway patency through bron-
weakness may occur somewhat less frequently. choscopic debridement of tumor, photodynamic
Physical examination may elicit signs of advanced therapy, airway stenting, or brachytherapy.
disease including the following: lymphadenopathy in The staging system for NSCLC is based on
the supraclavicular or cervical regions, percussion TNM classification. The 7th edition of the
Lung Cancer Surgery 105

• Separate nodule in an ipsilateral lobe (M1) has been


downstaged to T4.
TMN N0 N1 N2 N3
• M1 has been subdivided into M1a and M1b.
• Malignant pericardial or pleural effusion (T4) has
been upstaged to M1a.
T1a IA IIA IIIA IIIB
• Separate nodule in a contralateral lobe (M1) is
subclassified as M1a.
T1b IA IIA IIIA IIIB
• Distant metastasis is now designated as M1b.

2.1.1.1 T Stage
T1 tumors are \3 cm in diameter, surrounded com-
T2a IB IIA IIIA IIIB
pletely by lung parenchyma or visceral pleura and
exhibit invasion no more proximal than a lobar
bronchus. T2 tumors are[3 cm but limited to 7 cm in
T2b IIA IIB IIIA IIIB
diameter, can exhibit invasion of the visceral, but not
parietal pleura, and may involve the mainstem
bronchus no closer than within 2 cm of the carina.
T3 IIB IIIA IIIA IIIB
Atelectasis or obstructive pneumonitis may extend to
the hilum, but not involve an entire lung.
T3 tumors are [7 cm in diameter or exhibit locally
T4 IIIA IIIA IIIB IIIB invasive behavior, involving the parietal pleura, chest
wall (including the superior sulcus), diaphragm, phrenic
nerve, mediastinal pleura, parietal pericardium or a
M1a IV IV IV IV mainstem bronchus\2 cm from the carina. There may
be atelectasis or obstructive pneumonitis involving the
entire lung. In addition, T3 also describes satellite tumor
M1b IV IV IV IV
nodules within the same lobe. T4 tumors invade the
carina, trachea, mediastinum, vertebral body, recurrent
laryngeal nerve, great vessels, heart, or esophagus. If a
Fig. 1 Non-small cell lung cancer staging. 7th edition of
IALSC Staging System separate tumor nodule was present in an ipsilateral lobe,
this would also be considered a T4 lesion.
Chest CT is considered a mainstay for staging
International Lung Cancer Staging System was despite historical data that demonstrate an inability to
published in 2009 (Fig. 1) (Detterbeck et al. 2009), distinguish between T1/T2 and T3/T4 tumors up to
and subsequently adopted by the American Joint one-quarter of the time, (Antoch et al. 2003; Lardinois
Committee on Cancer (AJCC) and the Union et al. 2003) a limitation that may affect the extent of
Internationale Contre le Cancer (UICC) in 2010 surgical resection (Webb et al. 1991). Integrated
(Edge et al. 2010). While the N descriptor remained PET-CT scan may prove more accurate, yielding 98%
unaltered, the T (Rami-Porta et al. 2007) and M correct clinical staging when compared to pathologi-
(Postmus et al. 2007) descriptor classifications have cal staging (Lardinois et al. 2003). Radiologic
been revised extensively to provide greater prog- reconstruction of the tracheobronchial tree is insuffi-
nostic specificity: cient to exclude airway invasion, therefore, our
• T1 (B3 cm) has been subclassified into T1a policy is to perform bronchoscopy to assess the air-
(B2 cm) and T1b ([2 but B3 cm). way before surgical resection. Determination of the
• T2 ([3 cm) has been subclassified into T2a proximal extent of endobronchial tumor invasion
([3 but B5 cm) and T2b ([5 but B7 cm). (i.e., distance from carina) may be accomplished, in
• T2 ([7 cm) has been upstaged to T3. addition to assessment for anatomic abnormalities,
• Separate nodule in the same lobe (T4) has been which might influence the surgical resection. Intra-
downstaged to T3. operatively, frozen section analysis may be useful to
106 S. P. Gangadharan et al.

Fig. 2 The International


Association for the Study of
Lung Cancer (IASLC) lymph
node map (Rusch et al. 2009)

determine extrapulmonary tumor involvement versus Mountain–Dresler modification of the ATS map) was
inflammation or adhesion in some cases (i.e., confirm agreed upon by the AJCC and the UICC for the 6th
T3 or T4 status). edition of the staging manual (Mountain and Dresler
1997). N0 cancers have no demonstrable metastases
2.1.1.2 N Stage to regional lymph nodes. N1 represents metastasis to
The lymph node drainage of the lung has been the ipsilateral peribronchial or hilar lymph nodes, as
described previously (Asamura et al. 1999; Naruke well as direct extension of the primary tumor into
et al. 1978). In 1997, a revised lymph node map (the intrapulmonary nodes (lymph nodes with double-digit
Lung Cancer Surgery 107

numbering). N2 lymph nodes are metastases to ipsi- respectively, whereas those of PET/CT were 85, 84,
lateral mediastinal or subcarinal lymph nodes (single- and 84%, respectively (Shim et al. 2005). In practice,
digit lymph nodes). N3 designates metastasis to in cases with very small solitary peripheral tumors
contralateral mediastinal or hilar lymph nodes, or any and a negative mediastinum by PET-CT, invasive
scalene or supraclavicular lymph nodes irrespective staging may sometimes be omitted prior to surgical
of laterality. resection.
For the 7th edition of the staging manual, the Invasive lymph node staging can be accomplished
IASLC validated the prognostic influence of the by transthoracic needle biopsy, EUS needle biopsy,
existing N descriptor as previously defined. There mediastinoscopy, thoracoscopy, or more recently,
were, however, some discrepancies between this EBUS with trans-bronchial needle aspiration (EBUS–
nodal map and the one used in Asia. To address this TBNA). Meta-analysis of the first four techniques
and another concern about the reproducibility of reveals that cervical mediastinoscopy yields the best
determining the boundaries between different nodal performance profile, with a sensitivity of 81% and
stations during mediastinoscopy or thoracotomy negative predictive value of 91% (Toloza et al. 2003b).
(Zielinski and Rami-Porta 2007), the IASLC has Half of the nodes missed were not accessible through
developed a new lymph node map that defines six cervical mediastinoscopy, because this technique only
nodal ‘‘zones’’ encompassing the established nodal permits evaluation of the paratracheal and subcarinal
stations (Fig. 2) (Rusch et al. 2009). lymph node stations. Further enhancement of sensi-
Non-invasive lymph node staging can be per- tivity may be accomplished with the addition of
formed using CT scan, PET scan, MRI, endoscopic extended cervical or anterior mediastinotomy tech-
ultrasound (EUS), or endobronchial ultrasound niques (Ginsberg et al. 1987; McNeill and Chamberlain
(EBUS). A meta-analysis (Toloza et al. 2003a) of 1966). EBUS-TBNA has expanded the options for
studies utilizing the first four of these various staging NSCLC, extending the ability to sample sta-
modalities revealed a pooled sensitivity of 57% for tions to the hilar and even interlobar levels. In addition,
CT, 84% for PET, 100% for MRI, and 78% for EUS. its use for obtaining tissue from the primary lesion has
Specificities were 89% for CT, 82–95% for PET, 91% reduced the need for transthoracic biopsy and its risk
for MRI, and 71% for EUS. Subsequent assessment of for iatrogenic pneumothorax (5–8 vs. 23–38%) (Eber-
the ability of MRI to stage the mediastinum have been hardt et al. 2007). In patients found to have enlarged
equivocal (Webb et al. 1991). MRI is superior to CT nodes on CT, the sensitivity and specificity can be as
for characterizing direct invasion into the mediasti- high as 94 and 100%, respectively (Herth et al. 2006a).
num, chest wall, diaphragm, or vertebral bodies due When the CT was negative, results dropped off only
to its ability to detect differences in signal intensity slightly from 92.3 to 100%, with a negative predictive
between tumor and normal tissues, including bone, value of 96.3% (Herth et al. 2006b). A recent ran-
fat, vascular structures, and, soft tissue (Shiotani et al. domized controlled trial comparing mediastinoscopy
2000). It greatly facilitates the evaluation of superior versus EUS/EBUS plus mediastinoscopy found that the
sulcus tumors, particularly when assessing involve- addition of EUS/EBUS to mediastinoscopy improved
ment of the neural foramina or tumors abutting the sensitivity from 79 to 94% (Annema et al. 2010).
mediastinum, structures of the chest wall, or dia- A secondary endpoint of this study noted a trend toward
phragm. Currently, MRI would not be considered the improved sensitivity of 85% for EUS/EBUS alone,
standard imaging modality for evaluation of lymph with similar NPV as mediastinoscopy. Nevertheless, a
nodes in lung cancer. The accuracy of CT/PET scan negative result obtained by EBUS or EUS should still
with regard to lymph node staging may be enhanced prompt confirmation of true negativity via mediasti-
by the integration of these two technologies (Weng noscopy sampling.
et al. 2000). We routinely use integrated CT/PET to With the development of video-assisted medias-
stage individuals with enlarged nodes or patients tinoscopy (VAM), further improvements to what is
whose primary tumor demonstrates characteristics already considered the gold standard for staging the
concerning for nodal spread. In a prospective com- mediastinum were obtained. One report on the use of
parison of CT versus CT/PET, the sensitivity, speci- VAM to stage NSCLC in patients with enlarged
ficity, and accuracy of CT were 70, 69, and 69%, nodes by CT scan demonstrated a sensitivity of
108 S. P. Gangadharan et al.

97.3% and specificity 100% (Venissac et al. 2003). et al. 2003). Unnecessary thoracotomy, however, may
In cases of extensive tumor infiltration into the be prevented by routine extensive extrathoracic
mediastinum radiologic staging may suffice and workup (Anonymous 2001). In addition to head CT or
needle aspiration or bronchoscopy may be enough to MRI, we use whole body integrated PET-CT scanning
obtain pathologic confirmation of diagnosis (Kramer to further clarify the presence of metastatic disease;
and Groen 2003; Detterbeck et al. 2003). The utility the effect of this technology on survival and recur-
of thoracoscopic lymph node staging has not been rence is yet to be prospectively determined. Patients
fully elucidated (Gossot et al. 1996; Landreneau with either solitary brain metastases (Patchell et al.
et al. 1993a) but it would appear that VATS as an 1990; Burt et al. 1992; Magilligan et al. 1986) or
adjunct to mediastinoscopy is reasonable, especially solitary adrenal metastases (Luketich and Burt 1996;
for subaortic and para-aortic stations (Van Schil Porte et al. 2001) may benefit from surgical resection
2007). of the primary lung tumor in addition to the metastatic
When invasive LN sampling is indicated and con- lesion. Our strategy in these cases of ‘oligometastatic’
firmation of positive mediastinal nodal involvement is disease is to combine a metastatic workup with cer-
not achieved endoscopically, cervical mediastinoscopy vical mediastinoscopy. If contralateral nodal disease
is used as the last preoperative staging step before is not found, then the patient may undergo resection
planned surgical resection. To minimize the likelihood of the solitary brain or adrenal metastasis, followed
that lymph nodes will be read as falsely negative for by pulmonary resection as above.
tumor metastasis, we send the specimens for perma-
nent section analysis by pathology instead of relying
on frozen section analysis. Thus, the planned pul- 2.2 Preoperative Fitness
monary resection is deferred to a second operative
setting. The surgical approach to cervical mediasti- Once a patient is determined to be resectable, it is
noscopy and lymph node sampling commences with a imperative to assess overall fitness to undergo surgery.
small incision above the sternal notch, followed by In addition to a careful history and physical that might
dissection between the strap muscles until the pre-tra- reveal the presence of heart failure, coronary insuffi-
cheal fascia may be breached. Blunt dissection is then ciency, or other co-morbidities, all patients considered
used to enter the mediastinum, and the paratracheal for surgical resection at our institution undergo pul-
and subcarinal lymph nodes are exposed and removed monary function testing (PFT) and determination of the
with a biopsy forcep under direct vision (Reed and diffusing capacity of the lung for carbon monoxide
Sugarbaker 1996). The morbidity of the procedure is (DLCO). In addition, a modified stair-climbing test is
minimal (Park et al. 2003; Luke et al. 1986). Deter- sometimes used to assess a patient’s suitability for
mination of IIIB disease (contralateral mediastinal surgery (Brunelli et al. 2002, 2004).
lymph node involvement, N3) would preclude surgical A preoperative forced expiratory volume
resection. Our algorithm for positive N2 nodes (FEV1) [ 2L (60% of predicted) and DLCO [ 60%
involves preoperative chemoradiation or chemotherapy of predicted value suggest that the patient will tolerate
alone, prior to re-staging imaging. If there is no pro- pulmonary resection, including pneumonectomy.
gression of disease, we recommend pulmonary resec- The threshold to tolerate a lesser resection is com-
tion with radical lymphadenectomy. mensurately reduced: FEV1 [ 1L for lobectomy and
FEV1 [ 0.6L for wedge resection (Miller et al.
2.1.1.3 M Stage 1981). At our institution, we rely heavily on the
The absence of clinical findings may preclude the percent of predicted values, as these account for
need to scan the asymptomatic early-stage lung individual variability in age or size. Despite some
cancer patient extensively, as neither survival nor variance in the literature regarding the efficacy of
recurrence rates are affected (Tanaka et al. 1999; FEV1 or DLCO in predicting outcome after lung
Ichinose et al. 1989). Some authors recommended surgery (Stephan et al. 2000; Ferguson et al. 1988)
radiologic investigation for extrathoracic disease only further testing to stratify postoperative risk should be
if it is warranted by clinical evaluation or in the case undertaken if the FEV1 or DLCO is less than the
of advanced disease (Stages IIIA or IIIB) (Silvestri thresholds cited above (Datta and Lahiri 2003).
Lung Cancer Surgery 109

Calculation of a predicted postoperative FEV1 bronchopulmonary structures—pulmonary vein, pul-


(ppoFEV1) may be accomplished either by estimation, monary artery or bronchus—or the attendant draining
using the formula of Juhl and Frost [ppoFEV1 = pre- lymphatics or lymph nodes. Segmentectomy describes
operative FEV1 9 (1-(S 9 5.26)/100); S number of an anatomic resection of the bronchopulmonary seg-
segments to be resected] (Juhl and Frost 1975), or by ment. The right upper lobe is comprised of apical,
quantitative VQ scanning modified after Wernly et al. anterior, and posterior segments; the right middle lobe
(1980) [ppoFEV1 = preoperative FEV1 9 (1-(% is comprised of the lateral and medial segments; and
perfusion contributed by affected lung/100 9 S/total the right lower lobe is comprised of the superior
number of segments in affected lung))]. A ppo- segment, as well as the medial, anterior, lateral, and
FEV1 [ 40% of predicted and DLCO [ 40% of pre- posterior basal segments. The left upper lobe is
dicted have been suggested as threshold values (Datta divided into the upper division comprising the api-
and Lahiri 2003; Beckles et al. 2003). Our group has coposterior and anterior segments, and the lingula,
previously demonstrated that by using a variety of which contains a superior and inferior segment. The
minimally invasive techniques, limited resections, and left lower lobe is comprised of the superior segment,
concomitant lung volume reduction, with advanced and the anteromedial, lateral, and posterior basal
anesthetic and perioperative care, curative resections segments. Lobectomy entails removal of an entire lobe
can be performed in patients with preoperative and its lobar pulmonary artery, pulmonary vein, and
FEV1 \ 35% of predicted with a mortality of 1% and bronchus, with attendant lymphatic basin. Bilobec-
serious morbidity under 5% (Linden et al. 2005). The tomy is similar resection of two lobes from the same
determination of operability should be made by a tho- lung. Sleeve resection denotes the removal of a cir-
racic surgeon skilled in these techniques. Exercise test- cumferential portion of the airway in conjunction
ing and calculation of VO2max represents the next level of with the parenchymal resection. The remaining
testing should the predicted postoperative values lung requires a bronchial anastomosis in order to
be below threshold values. VO2max [ 20 ml/kg/min re-establish airway continuity. A sleeve resection can
designates an acceptable risk group for surgery. also be performed on the pulmonary artery, should it
VO2max \ 10 ml/kg/min confers significantly increased be necessary to resect a circumferential portion of the
risk for postoperative death or cardiopulmonary com- vessel with the specimen. Similarly, bronchoplasty or
plications following lung surgery (Datta and Lahiri arterioplasty in conjunction with a pulmonary resec-
2003; Beckles et al. 2003). tion describe the techniques by which the bronchus or
In patients who are scheduled to undergo pneumo- pulmonary artery is reconstructed after removal of a
nectomy or who may present with cardiac co-morbidity, non-circumferential portion of the structure during
we obtain a preoperative echocardiogram to evaluate resection. Pneumonectomy can be intrapericardial or
ventricular and valvular function, as well to investigate extrapericardial, in reference to the site of division of
any pre-existing pulmonary hypertension. Occasion- the pulmonary vessels. Extrapleural pneumonectomy
ally, right heart catheterization and pulmonary artery or pleuropneumonectomy refer to the en bloc removal
balloon occlusion are used to determine further a of the parietal pleura with the entire lung. En bloc chest
patient’s physiologic response to lung resection. wall resection describes the removal of a portion of the
parietal pleura, ribs, and intercostal musculature
attached to the primary specimen of resected lung.
3 Operative Strategy

3.1 Nomenclature and Anatomy 3.2 Extent of Resection

The scope of surgical resection ranges from wedge The first lobectomy for lung cancer using a technique
resection to pneumonectomy. The wedge resection of individual ligation of the hilar structures was per-
represents a non-anatomic resection of the target formed by Davies in 1912 (Davies 1913). Churchill
lesion, with a variable margin of lung parenchyma. refined lung resection with the introduction of the
The terminology non-anatomic refers to the lack of technique of individual ligation of the bronchopulmo-
dissection of any of the branches of the three nary structures (Churchill and Belsey 1939). Graham
110 S. P. Gangadharan et al.

reported the first successful pneumonectomy for lung et al. (2002) found that for T1 tumors \2 cm in
cancer in 1933 (Graham and Singer 1933). Pneumo- diameter, 5-year survival (87.1%) was nearly equiva-
nectomy remained the operation of choice for lung lent to lobectomy (87.8%). Schuchert and associates
cancer until Churchill’s report in 1950, which detailed from the University of Pittsburgh retrospectively
long-term survival following lobectomy (Churchill reviewed their experience with sublobar resection, both
et al. 1950). The issue of whether sublobar or non- segmentectomy and wedge resection, and found no
anatomic resection might similarly suffice was raised significant difference in recurrence or cancer-free sur-
by Jensik et al. (1973). Subsequent investigators have vival (Schuchert et al. 2011). For AIS, a Japanese study
concluded that a lesser resection in the setting of demonstrated wedge resection to have a 5-year overall
impaired cardiopulmonary reserve or advanced age survival rate and the disease-free survival rate were
might be justified (Landreneau et al. 1997; Errett et al. both 93%, and the 5-year cancer-specific survival rate
1985). One argument against applying a strategy of was 100% (Koike et al. 2009). The Cancer and
limited resection to any patient with early-stage lung Leukemia Group B (CALGB) 140503 trial, a ran-
cancer has been that it may understage cancers by domized study of lobectomy versus sublobar resection
virtue of inadequate lymph node sampling. Takizawa in patients with small peripheral Stage IA NSCLC, is
et al. (1998) found a 17% incidence of metastases to expected to have an enrollment of 1258 patients with
N1 and N2 lymph nodes with radical lymphadenec- the primary end point being a comparison of disease-
tomy after resection of small (1.1–2.0 cm) peripheral free survival (National Cancer Institute 2010). Until
lung adenocarcinomas, suggesting that an adequate the results of this trial are known, the controversy
assessment of the draining lymph node basin may be surrounding the issue will likely continue.
important even in these distal T1 tumors. The other At our institution, we reserve pneumonectomy for
area of contention is whether limited resection could cases in which tumors are too central to fully resect
effect a local and systemic cure. A randomized trial of with lobectomy, and where bronchoplasty or sleeve
lobectomy versus segmentectomy or wedge resection resection still would not allow an adequate margin with
for T1N0 NSCLC was reported by the Lung Cancer parenchymal sparing. Similarly, extensive involvement
Study Group in 1995. Although no statistically sig- of the pulmonary vessels may necessitate pneumo-
nificant difference in survival was found, they did find nectomy, if arterioplasty is not feasible. Also, in cases
an increased overall recurrence rate and locoregional where the cancer crosses the fissure on the left or
recurrence in the limited resection group (75% and crosses the major fissure, involving the upper and lower
threefold, respectively) (Ginsberg and Rubinstein lobes on the right, consideration is made to proceed
1995). This increased recurrence after sublobar resec- with pneumonectomy in patients with suitable reserve.
tion echoed the findings of Warren and Faber (1994). Conversely, limited resection for lung cancer, with
Landreneau et al. (1997) examined the outcomes of the exception of a pure AIS lesion, is reserved at our
lobectomy and wedge resection, both video-assisted institution for patients with marginal medical status,
and via open thoracotomy, and found a significant advanced age, poor pulmonary reserve, or in some
improvement in 5-year survival curves after lobec- instances of second primary lung cancer. In all cases the
tomy, although this was explicable by an excess of tumor stage is T1. Every effort is made to adequately
non-cancer-related deaths in the limited resection stratify risk preoperatively, to ensure that all potential
group. Miller and associates in their retrospective candidates for anatomic resection are identified.
analysis also found decreased 5-year survival for sub-
lobar resection compared to lobectomy. Importantly,
however, they also found that survival after segmen- 3.3 Technique of Resection
tectomy was statistically better then after wedge
resection, (57 and 27%, respectively), highlighting the We have previously described in detail the steps
principle of an anatomic segment as the functional for the major pulmonary resections (Sugarbaker
oncologic unit (Miller et al. 2002). et al. 2001). In brief, after the induction of general
Indeed, when sublobar resection consists of seg- anesthesia, bronchoscopy is performed to assess the
mentectomy with complete lobar and mediastinal dis- airway for unexpected tumor progression or anatomic
section (i.e., extended segmentectomy), Yoshikawa abnormality that would alter the planned resection.
Lung Cancer Surgery 111

Subcutaneous heparin and prophylactic antibiotics are et al. 1997). Myrdal et al. (2001) reviewed their
administered. Single-lung ventilation is then accom- experience of 616 patients undergoing lung cancer
plished using a double-lumen endotracheal tube or surgery and found an overall 30-day postoperative
single-lumen tube with a bronchial blocker. The mortality rate of 2.9%, with pneumonectomy con-
patient is positioned in thoracotomy position—a lat- ferring a higher risk (5.7%) than lobectomy (0.6%).
eral decubitus position, with the operative side up. The rate of major complication (defined as post-
A number of incisions may be used to access the operative bleeding leading to exploration, respira-
pleural space. For most anatomic resections, we utilize tory failure, bronchopleural fistula, myocardial
a posterolateral thoracotomy incision that begins at a infarction, stroke, heart failure, or renal failure) was
point midway between the lower half of the scapula and 8.8%, with a higher rate seen after pneumonectomy
the spine, and extends to the anterior border of the la- (18.5%) than lobectomy (5.7%). Minor events
tissimus dorsi muscle. The serratus muscle is usually occurred in 22%, with supraventricular arrhythmias
spared, the latissimus muscle is usually divided. The accounting for half of these complications. The
fifth intercostal space is entered at the superior border mortality and complication rates after bilobectomy
of the sixth rib. Occasionally the sixth space is used, or a have been previously reported to be comparable to
rib may be removed partially or entirely in order to that of pneumonectomy (Deneuville et al. 1992).
widen the access to the chest. An anterolateral thora- However, more recent series do not report excess
cotomy, usually in the fourth intercostal space, is mortality or morbidity after bilobectomy (Damhuis
another alternative. Sufficient analgesia for these inci- and Schutte 1996; Cerfolio et al. 2000; Carbognani
sions is achieved with the combination of a long-acting et al. 2001).
local anesthetic and a narcotic via a thoracic epidural At the Brigham and Women’s Hospital we use
catheter placed preoperatively. postoperative clinical pathways to standardize care
The hilar structures are individually dissected and after pulmonary resection and to reduce length of
divided. Our preference is to divide both vessels and stay. Implementation of patient care pathways for
bronchi using a stapler. Smaller pulmonary arterial lobectomy has been reported to reduce both length of
branches are doubly ligated if they are not amenable stay and hospital cost. Although 1-day stays after
to stapler division. Incomplete fissures are also divi- lobectomy have been reported, a length of stay in the
ded using a stapling device. Lymphadenectomy is 5-to-7 day range is more common (Cerfolio et al.
performed. 2001; Wright et al. 1997) Although 1-day stays after
The margins are inspected by a pathologist upon lobectomy have been reported, Tovar et al. (1998) a
removal of the specimen to assure a negative bron- length of stay in the 5-to-7 day range is more com-
chial margin. The integrity of the bronchial stump is mon (Tschernko et al. 1996; Kirby et al. 1995). At our
checked by testing the stump with ventilatory pres- institution all patients are transferred to a specialized
sure up to 30 cm of H2O. When the patient has thoracic surgery intermediate care unit after lobec-
received neoadjuvant radiation or may receive tomy or lesser resection. After 1-to-3 days in this
postoperative radiation, it is our preference to but- setting, to permit invasive hemodynamic monitoring,
tress the bronchial stump with an intercostal muscle continuous oxygen monitoring, and frequent pul-
pedicle, a pericardial or pleural flap, or a thymic fat monary toilet/ambulation, patients are transferred to a
pad. We also buttress after pneumonectomy or regular floor bed on the thoracic surgical unit, which
bilobectomy. provides continued specialized nursing care. After
pneumonectomy, our patients are recovered first in a
specialized thoracic intensive care unit, which allows
3.4 Postoperative Course even more extensive monitoring such as pulmonary
artery catheterization, if indicated. Both hospital and
Mortality for lung cancer surgery ranges from 2 to 4% surgeon-specific experience influence postoperative
in modern series, with postoperative morbidity mortality for lobectomy and pneumonectomy, with
occurring approximately 15–30% of the time higher volume correlating with better outcomes in
(Deslauriers et al. 1989; Ginsberg et al. 1983; several large studies (Birkmeyer et al. 2002, 2003;
Knott-Craig et al. 1997; Myrdal et al. 2001; Yano Hannan et al. 2002).
112 S. P. Gangadharan et al.

Five-year survival for NSCLC has been reported endoscopic bag to avoid seeding the port sites with
by Goldstraw in 2007 (Goldstraw et al. 2007). For shed tumor cells.
Stage IA tumors the 5-year survival is 73% (T1N0) The operative mortality for wedge resection for
and 58% for Stage IB (T2N0). Stage IIA tumors are lung cancer has been reported to be negligible
46% (T1N1, T2aN1, T2bN0), 36% for Stage IIB (Landreneau et al. 1997; Kodama et al. 1997). For
(T2bN1, T3N0). In Stage IIIA, the 5-year survival is elderly patients undergoing VATS wedge resection,
24% (T1-2N2, T3N1-2, T4N0-1). The rate drops in we have previously shown that the mortality is \1%,
Stage IIIB (T4N2, T1-4N3) to 9%. For Stage IV with a morbidity of 9%. VATS lobectomy also has
(TxNxM1-2) overall 5-year survival is 13%. been accomplished with minimal mortality and mor-
bidity (Gharagozloo et al. 2003; Tatsumi and Ueda
2003; Morgan et al. 2003; Lewis et al. 1999;
3.5 Video-Assisted Thoracoscopic McKenna 1998). A recent representative report by
Surgery Walker and colleagues describes their experience
with 158 patients undergoing VATS lobectomy. They
Video-assisted thoracoscopic surgery (VATS) utilizes report an 11% rate of conversion to open thoracot-
small port accesses to the chest and a videoscope for omy, secondary to extent of disease and bleeding in
visualization, thereby avoiding a full thoracotomy most cases. The in-hospital mortality rate was 0.6%,
incision. The ability to perform pulmonary resections with an overall 30-day mortality rate of 1.8%
with VATS techniques has provided a less invasive (Morgan et al. 2003). In a series of over 1,000 VATS
method to safely diagnose and treat lung cancers lobectomy cases, McKenna reported no intraoperative
(DeCamp et al. 1995). Initial videoscopic thoracic deaths, and a 30-day mortality rate of just 0.9%, none
surgery was diagnostic or limited to treatment of of which was secondary to bleeding. The rate of
pneumothorax, pleural effusion, or other benign con- conversion to open thoracotomy was 2.5% (McKenna
ditions (Kopp et al. 1979; Oldenburg and Newhouse et al. 2006).
1979; Rodgers et al. 1979; Kapsenberg 1981; Boutin The oncologic validity of VATS lobectomy has not
et al. 1982; Fritsch et al. 1975). VATS lobectomies been addressed in a randomized prospective trial, but
for lung cancer were first reported in 1993 (Walker retrospective studies report 5-year survival rates for
et al. 1993; Kirby and Rice 1993). Stage I and II NSCLC ranging from 60 to 90%, and
We have described our technique of VATS locoregional recurrence rates around 5% (Tatsumi
lobectomy in detail elsewhere (Sugarbaker et al. and Ueda 2003; Walker et al. 2003; Sugi et al. 2000;
2001). A small incision in the anterior seventh inter- McKenna et al. 1998). Freedom from cancer-related
space is used to place a port for a 5- or 10-mm vid- or associated death has been reported to be 78% for
eoscope. The entirety of the pleural space and lung Stage I cancers, 51% for Stage II, and 29% for Stage
may be inspected for unexpected local or metastatic III (Walker et al. 2003). An adequate lymph node
spread. For wedge resections, second and third ports dissection appears to be possible during VATS
are placed so that a triangulation is achieved over the lobectomy (Asamura et al. 1999; Morikawa et al.
tumor, and instruments can be introduced to retract, 1998).
dissect, and staple the lung. For formal lobectomies or An initial randomized trial of VATS versus open
segmentectomies we use a 4 cm fourth interspace lobectomy did not demonstrate a statistically signifi-
accessory incision in the anterior axillary line that cant difference in length of stay (Kirby et al. 1995).
allows access to the hilar structures. No rib spreading A subsequent randomized trial and several non-
is done. In addition, we employ one posterior 1.5 cm randomized trials, however, were able to show a
port near the tip of the scapula for retraction and an reduction in length of stay with the minimally inva-
additional small port anteriorly for suction. Dissection sive technique (Tschernko et al. 1996; Demmy and
and division of the pulmonary vein, pulmonary artery, Curtis 1999; Ohbuchi et al. 1998). These same trials
and bronchial structures are accomplished with have also reported a significant difference in the level
endoscopic staplers in similar manner to our open of pain associated with VATS lobectomy (Tschernko
lobectomy. Mediastinal lymphadenectomy is also et al. 1996; Morikawa et al. 1998; Demmy and Curtis
performed. The specimen is removed via an 1999; Landreneau et al. 1993b). Walker and
Lung Cancer Surgery 113

associates have shown that the incidence of chronic were initially approached with preoperative radiother-
pain following VATS lobectomy is 1.2% (Walker apy alone (Shaw et al. 1961). Recently, however, ret-
et al. 1996). A recent review (Rueth and Andrade rospective studies (Wright et al. 2002; Attar et al. 1998)
2010) of VATS lobectomy versus open incorporated as well as a prospective randomized trial (Martin et al.
the findings from several large studies on the out- 2001) have shown potential benefit to combining che-
comes from open (Allen et al. 2006; Boffa et al. 2008) motherapy with preoperative radiation for superior
and VATS lobectomy (Flores et al. 2009; Swanson sulcus tumors.
et al. 2007). The data strongly support the oncologic Surgical approaches to superior sulcus tumors
equivalence of VATS compared to open lobectomy include extended posterolateral thoracotomy and
for patients with early-stage NSCLC. VATS was also anterior cervicothoracic incisions (Shaw et al. 1961;
associated with fewer postoperative complications Dartevelle et al. 1993). Resection usually comprises
and may have less of a negative biologic impact. the following steps: (1) resection of the chest wall
including first rib and, at times, portions of involved
vertebral bodies; (2) resection of involved nerve roots,
3.6 Radiation Therapy for Patients up to the first thoracic nerve root; (3) resection of the
Undergoing Lung Resection: T3 thoracic sympathetic chain; (4) resection of upper
Tumors lobe or wedge of involved lung; and (5) lymph node
dissection. Incomplete resection yields a survival rate
Adjuvant and neoadjuvant chemotherapy with or which is comparable to that of no resection (Rusch
without radiation has been studied extensively for et al. 2000).
Stage IIIA (N2) and Stage IIIB (N3) disease. T3 tumors involving the mediastinum are very
Radiation therapy, without chemotherapy, does little difficult to cure. Burt et al. (1987) reviewed 225
to help these patients. For locally invasive, T3, patients accrued over an 11-year period at Memorial
tumors, radiation therapy alone may make the dif- Sloan Kettering. The 5-year survival for patients with
ference between clear margins and positive margins. T3N2 disease was 8%, which is similar to survival of
In general, T3 tumors invading the chest wall are patients with lower T stage tumors and N2 disease.
treated with surgical excision with wide margins Patients with T3N0 tumors invading the mediastinum
alone, without the need for radiation therapy. Sev- fared no better, with 5-year survival of 10%.
eral retrospective trials have shown either no benefit, Although prospective trials do not exist, this subset of
or a detriment to adding preoperative radiation patients may very well benefit from neoadjuvant
therapy to patients with simple chest wall invasion radiation or chemoradiation.
(Piehler et al. 1982; Albertucci et al. 1992).
Exceptions to this finding are tumors abutting or
involving the vertebral bodies. If the tumor is close References
to the vertebral body, one may consider preoperative
radiation in order to shrink the tumor and lessen the Albertucci M, DeMeester TR, Rothberg M et al (1992) Surgery
chance that resection of the vertebral body will be and the management of peripheral lung tumors adherent to
required. the parietal pleura. J Thorac Cardiovasc Surg 103:8–13
Allen MS, Darling GE, Pechet TTV et al (2006) Morbidity and
Superior sulcus tumors (Pancoast tumors) represent mortality of major pulmonary resections in patients with
a unique subset of tumors that invade the chest apex. early-stage lung cancer: initial results of the randomized,
The tumor may involve vertebral bodies, subclavian prospective ACOSOG Z0030 trial. Ann Thorac Surg
vessels, or the brachial plexus. Pancoast tumors are 81:1013–1020
American Cancer Society. Cancer Facts & Figures 2010.
preoperatively staged by CT, and sometimes MRI, as Atlanta: American Cancer Society; 2010
T3 or T4 depending on the level of invasion. Lymph Annema JT, van Meerbeeck JP, Rintoul RC et al (2010)
node staging and metastatic workup are undertaken as Mediastinoscopy vs endosonography for mediastinal nodal
for other NSCLC. Positive N2 and N3 nodes are asso- staging of lung cancer. JAMA 304:2245–2252
Anonymous (2001) Investigating extrathoracic metastatic
ciated with a 5-year survival of \10% (Detterbeck disease in patients with apparently operable lung cancer.
1997; Deslauriers et al. 1994). Treatment of Pancoast The Canadian Lung Oncology Group. Ann Thorac Surg
tumors begins with neoadjuvant therapy. These tumors 71:425–433 (discussion 433–434)
114 S. P. Gangadharan et al.

Antoch G, Stattaus J, Nemat AT et al (2003) Non-small cell cases treated at the Massachusetts General Hospital from
lung cancer: dual-modality PET/CT in preoperative staging. 1930 to 1950. J Thorac Cardiovasc Surg 20:349–365
Radiology 229:526–533 Cummings SR, Lillington GA, Richard RJ (1986) Estimating the
Asamura H, Nakayama H, Kondo H, Tsuchiya R, Naruke T probability of malignancy in solitary pulmonary nodules.
(1999) Lobe-specific extent of systematic lymph node A Bayesian approach. Am Rev Respir Dis 134:449–452
dissection for non-small cell lung carcinomas according to Damhuis RA, Schutte PR (1996) Resection rates and post-
a retrospective study of metastasis and prognosis. J Thorac operative mortality in 7, 899 patients with lung cancer. Eur
Cardiovasc Surg 117:1102–1111 Respir J 9:7–10
Attar S, Krasna MJ, Sonett JR et al (1998) Superior sulcus Dartevelle PG, Chapelier AR, Macchiarini P et al (1993)
(Pancoast) tumor: experience with 105 patients. Ann Thorac Anterior transcervical-thoracic approach for radical resec-
Surg 66:193–198 tion of lung tumors invading the thoracic inlet. J Thorac
Beckles MA, Spiro SG, Colice GL, Rudd RM, American Cardiovasc Surg 105:1025–1034
College of Chest Physicians (2003) The physiologic Datta D, Lahiri B (2003) Preoperative evaluation of patients
evaluation of patients with lung cancer being considered undergoing lung resection surgery. Chest 123:2096–2103
for resectional surgery. Chest 123:105S–114S Davies HM (1913) Recent advances in the surgery of the lung
Birkmeyer JD, Siewers AE, Finlayson EV et al (2002) Hospital and pleura. Br J Surg 1:228–231
volume and surgical mortality in the United States. N Engl J DeCamp MM Jr, Jaklitsch MT, Mentzer SJ, Harpole DH Jr,
Med 346:1128–1137 Sugarbaker DJ (1995) The safety and versatility of video-
Birkmeyer JD, Stukel TA, Siewers AE, Goodney PP, Wenn- thoracoscopy: a prospective analysis of 895 consecutive
berg DE, Lucas FL (2003) Surgeon volume and operative cases. J Am Coll Surg 181:113–120
mortality in the United States. N Engl J Med Demmy TL, Curtis JJ (1999) Minimally invasive lobectomy
349:2117–2127 directed toward frail and high-risk patients: a case-control
Boffa DJ, Allen MS, Grab JD, Gaissert HA, Harpole DH, study. Ann Thorac Surg 68:194–200
Wright CD (2008) Data from the society of thoracic Deneuville M, Regnard JF, Coggia M, Rojas-Miranda A,
surgeons general thoracic surgery database: the surgical Dartevelle P, Levasseur P (1992) The place for bilobectomy
management of primary lung tumors. J Thorac Cardiovasc in bronchogenic carcinoma. Eur J Cardiothorac Surg 6:
Surg 135:247–254 446–451
Boutin C, Viallat JR, Cargnino P, Rey F (1982) Thoracoscopic Deslauriers J, Ginsberg RJ, Dubois P, Beaulieu M, Goldberg M,
lung biopsy. Experimental and clinical preliminary study. Piraux M (1989) Current operative morbidity associated
Chest 82:44–48 with elective surgical resection for lung cancer. Can J Surg
Boyle P, Levin B (2008) International agency for research on 32:335–339
cancer. World cancer report 2008. IARC Press, Lyon Deslauriers J, Ginsberg RJ, Piantadosi S, Fournier B (1994)
Brunelli A, Al Refai M, Monteverde M, Borri A, Salati M, Prospective assessment of 30-day operative morbidity for
Fianchini A (2002) Stair climbing test predicts cardiopul- surgical resections in lung cancer. Chest 106:329S–330S
monary complications after lung resection. Chest Detterbeck FC (1997) Pancoast (superior sulcus) tumors. Ann
121:1106–1110 Thorac Surg 63:1810–1818
Brunelli A, Monteverde M, Al Refai M, Fianchini A (2004) Detterbeck FC, DeCamp MM Jr, Kohman LJ, Silvestri GA
Stair climbing test as a predictor of cardiopulmonary (2003) Lung cancer. Invasive staging: the guidelines. Chest
complications after pulmonary lobectomy in the elderly. 123:167S–175S
Ann Thorac Surg 77:266–270 Detterbeck FC, Boffa DJ, Tanoue LT (2009) The new lung
Burt M, Pomerantz A, Bains MS et al (1987) Results of surgical cancer staging system. Chest 136:260–271
treatment of stage III lung cancer invading the mediastinum. Eberhardt R, Anantham D, Ernst A, Feller-Kopman D, Herth F
Surg Clin N Am 67:987–1000 (2007) Multimodality bronchoscopic diagnosis of peripheral
Burt M, Wronski M, Arbit E, Galicich JH (1992) Resection of lung lesions: a randomized controlled trial. Am J Respir Crit
brain metastases from non-small-cell lung carcinoma. Care Med 176:36–41
Results of therapy. Memorial Sloan-Kettering Cancer Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti
Center Thoracic Surgical Staff. J Thorac Cardiovasc Surg A (2010) The American Joint Committee Cancer Staging
103:399–410 (discussion 410–411) Handbook. 7th edition, Springer Science and Business
Carbognani P, Tincani G, Solli P et al (2001) The bilobecto- Media, New York, pp 253–270
mies for lung cancer. J Cardiovasc Surg 42:421–424 Errett LE, Wilson J, Chiu RC, Munro DD (1985) Wedge
Cerfolio RJ, Holman WL, Katholi CR (2000) Pneumoperito- resection as an alternative procedure for peripheral
neum after concomitant resection of the right middle and bronchogenic carcinoma in poor-risk patients. J Thorac
lower lobes (bilobectomy). Ann Thorac Surg 70:942–946 Cardiovasc Surg 90:656–661
(discussion 6–7) Ferguson MK, Little L, Rizzo L et al (1988) Diffusing capacity
Cerfolio RJ, Pickens A, Bass C, Katholi C (2001) Fast-tracking predicts morbidity and mortality after pulmonary resection.
pulmonary resections. J Thorac Cardiovasc Surg 122:318–324 J Thorac Cardiovasc Surg 96:894–900
Churchill ED, Belsey HR (1939) Segmental pneumonectomy in Flores RM, Park BJ, Dycoco J et al (2009) Lobectomy by
bronchiectasis: lingula segment of the upper lobe. Ann Surg video-assisted thoracic surgery (VATS) versus thoracotomy
109:481 for lung cancer. J Thorac Cardiovasc Surg 138:11–18
Churchill ED, Sweet RH, Sutter L, Scannel JG (1950) The Fritsch A, Kokoschka R, Mach K (1975) Results of thoraco-
surgical management of carcinoma of the lung: a study of scopic sympathectomy in hyperhidrosis of the upper
Lung Cancer Surgery 115

extremities (author’s transl). Wien Klin Wochenschr Kapsenberg PD (1981) Thoracoscopic biopsy under visual
87:548–550 control. Poumon Coeur 37:313–316
Gharagozloo F, Tempesta B, Margolis M, Alexander EP (2003) Kirby TJ, Rice TW (1993) Video-assisted pulmonary lobect-
Video-assisted thoracic surgery lobectomy for stage I lung omy. Semin Thorac Cardiovasc Surg 5:316–320
cancer. Ann Thorac Surg 76:1009–1014 (discussion 14–15) Kirby TJ, Mack MJ, Landreneau RJ, Rice TW (1995)
Ginsberg RJ, Rubinstein LV (1995) Randomized trial of Lobectomy-video-assisted thoracic surgery versus muscle-
lobectomy versus limited resection for T1 N0 non-small sparing thoracotomy. A randomized trial. J Thorac Cardio-
cell lung cancer. Lung Cancer Study Group. Ann Thorac vasc Surg 109:997–1001 (discussion -2)
Surg 60:615–622 (discussion 622–623) Knott-Craig CJ, Howell CE, Parsons BD, Paulsen SM, Brown
Ginsberg RJ, Hill LD, Eagan RT et al (1983) Modern thirty-day BR, Elkins RC (1997) Improved results in the management
operative mortality for surgical resections in lung cancer. of surgical candidates with lung cancer. Ann Thorac Surg
J Thorac Cardiovasc Surg 86:654–658 63:1405–1409 (discussion 1409–1410)
Ginsberg RJ, Rice TW, Goldberg M, Waters PF, Schmocker BJ Kodama K, Doi O, Higashiyama M, Yokouchi H (1997)
(1987) Extended cervical mediastinoscopy. A single staging Intentional limited resection for selected patients with T1
procedure for bronchogenic carcinoma of the left upper N0 M0 non-small-cell lung cancer: a single-institution
lobe. J Thorac Cardiovasc Surg 94:673–678 study. J Thorac Cardiovasc Surg 114:347–353
Goldstraw P, Crowley J, Chansky K et al (2007) The IASLC lung Koike T, Togashi K, Shirato T et al (2009) Limited resection
cancer staging project: proposals for the revision of the tnm for noninvasive bronchioloalveolar carcinoma diagnosed by
stage groupings in the forthcoming (seventh) edition of the intraoperative pathologic examination. Ann Thorac Surg
TNM classification of malignant tumours. Journal of thoracic 88:1106–1111
oncology 2:706–714. doi:10.1097/JTO.0b013e31812f3c1a Kopp C, Perruchoud A, Schwander R, Herzog H (1979)
Gossot D, Toledo L, Fritsch S, Celerier M (1996) Mediasti- Thorascopy as a diagnostic and therapeutic precaution in
noscopy vs thoracoscopy for mediastinal biopsy. Results of lung and pleural diseases. Schweiz Med Wochenschr
a prospective nonrandomized study. Chest 110:1328–1331 109:478–480
Graham EA, Singer JJ (1933) Successful removal of the entire Kramer H, Groen HJ (2003) Current concepts in the
lung for carcinoma of the bronchus. J Am Med Assoc mediastinal lymph node staging of nonsmall cell lung
101:1371–1374 cancer. Ann Surg 238:180–188
Gurney JW (1993) Determining the likelihood of malignancy in Landreneau RJ, Hazelrigg SR, Mack MJ et al (1993a)
solitary pulmonary nodules with Bayesian analysis. Part I. Thoracoscopic mediastinal lymph node sampling: useful
Theory. Radiology 186:405–413 for mediastinal lymph node stations inaccessible by cervical
Gurney JW, Lyddon DM, McKay JA (1993) Determining the mediastinoscopy. J Thorac Cardiovasc Surg 106:554–558
likelihood of malignancy in solitary pulmonary nodules Landreneau RJ, Hazelrigg SR, Mack MJ et al (1993b)
with Bayesian analysis. Part II. Application. Radiology Postoperative pain-related morbidity: video-assisted tho-
186:415–422 racic surgery versus thoracotomy. Ann Thorac Surg
Hannan EL, Radzyner M, Rubin D, Dougherty J, Brennan MF 56:1285–1289
(2002) The influence of hospital and surgeon volume on in- Landreneau RJ, Sugarbaker DJ, Mack MJ et al (1997) Wedge
hospital mortality for colectomy, gastrectomy, and lung resection versus lobectomy for stage I (T1 N0 M0) non-
lobectomy in patients with cancer. Surgery 131:6–15 small-cell lung cancer. J Thorac Cardiovasc Surg
Herth FJ, Eberhardt R, Vilmann P, Krasnik M, Ernst A (2006a) 113:691–698 (discussion 698–700)
Real-time endobronchial ultrasound guided transbronchial Lardinois D, Weder W, Hany TF et al (2003) Staging of non-
needle aspiration for sampling mediastinal lymph nodes. small-cell lung cancer with integrated positron-emission
Thorax 61:795–798 tomography and computed tomography. N Engl J Med
Herth FJ, Ernst A, Eberhardt R, Vilmann P, Dienemann H, 348:2500–2507
Krasnik M (2006b) Endobronchial ultrasound-guided trans- Lewis RJ, Caccavale RJ, Bocage JP, Widmann MD (1999)
bronchial needle aspiration of lymph nodes in the radio- Video-assisted thoracic surgical non-rib spreading simulta-
logically normal mediastinum. Eur Respir J 28:910–914 neously stapled lobectomy: a more patient-friendly oncolo-
Howlader N, Noone A, Krapcho M (eds) (2011) SEER cancer gic resection. Chest 116:1119–1124
statistics review, 1975–2008. National Cancer Institute, Linden PA, Bueno R, Colson YL et al (2005) Lung resection in
Bethesda patients with preoperative FEV1 \ 35% predicted. Chest
Ichinose Y, Hara N, Ohta M et al (1989) Preoperative 127:1984–1990
examination to detect distant metastasis is not advocated Luke WP, Pearson FG, Todd TR, Patterson GA, Cooper JD
for asymptomatic patients with stages 1 and 2 non-small cell (1986) Prospective evaluation of mediastinoscopy for
lung cancer. Preoperative examination for lung cancer. assessment of carcinoma of the lung. J Thorac Cardiovasc
Chest 96:1104–1109 Surg 91:53–56
Jensik RJ, Faber LP, Milloy FJ, Monson DO (1973) Segmental Luketich JD, Burt ME (1996) Does resection of adrenal
resection for lung cancer. A fifteen-year experience. J Tho- metastases from non-small cell lung cancer improve
rac Cardiovasc Surg 66:563–572 survival? Ann Thorac Surg 62:1614–1616
Juhl B, Frost N (1975) A comparison between measured and Magilligan DJ Jr, Duvernoy C, Malik G, Lewis JW Jr,
calculated changes in the lung function after operation for Knighton R, Ausman JI (1986) Surgical approach to lung
pulmonary cancer. Acta Anaesthesiol Scand Suppl cancer with solitary cerebral metastasis: twenty-five years’
57:39–45 experience. Ann Thorac Surg 42:360–364
116 S. P. Gangadharan et al.

Martin J, Ginsberg RJ, Abolhoda A et al (2001) Morbidity and Porte H, Siat J, Guibert B et al (2001) Resection of adrenal
mortality after neoadjuvant therapy for lung cancer: the metastases from non-small cell lung cancer: a multicenter
risks of right pneumonectomy. Ann Thorac Surg study. Ann Thorac Surg 71:981–985
72:1149–1154 Postmus PE, Brambilla E, Chansky K et al (2007) The IASLC
McKenna RJ, Jr (1998) The current status of video-assisted lung cancer staging project: proposals for revision of the M
thoracic surgery lobectomy. Chest Surg Clin N Am descriptors in the forthcoming (seventh) edition of the TNM
8:775–85, viii, (discussion 87-88) classification of lung cancer. J Thorac Oncol 2:686–693
McKenna RJ Jr, Wolf RK, Brenner M, Fischel RJ, Wurnig P Rami-Porta R, Ball D, Crowley J et al (2007) The IASLC lung
(1998) Is lobectomy by video-assisted thoracic surgery an cancer staging project: proposals for the revision of the T
adequate cancer operation? Ann Thorac Surg 66:1903–1908 descriptors in the forthcoming (seventh) edition of the TNM
McKenna RJ Jr, Houck W, Fuller CB (2006) Video-assisted classification for lung cancer. J Thorac Oncol 2:593–602
thoracic surgery lobectomy: experience with 1, 100 cases. Reed MF, Sugarbaker DJ (1996) Mediastinal staging of lung
Ann Thorac Surg 81:421–426 cancer. Lippincott-Raven, Philadelphia
McNeill TM, Chamberlain JM (1966) Diagnostic anterior Rodgers BM, Moazam F, Talbert JL (1979) Thoracoscopy.
mediastinotomy. Ann Thorac Surg 2:532–539 Early diagnosis of interstitial pneumonitis in the immuno-
Miller JI, Grossman GD, Hatcher CR (1981) Pulmonary logically suppressed child. Chest 75:126–130
function test criteria for operability and pulmonary resec- Rueth NM, Andrade RS (2010) Is VATS lobectomy better:
tion. Surg Gynecol Obstet 153:893–895 perioperatively, biologically and oncologically? Ann
Miller DL, Rowland CM, Deschamps C, Allen MS, Trastek VF, Thorac Surg 89:S2107–S2111
Pairolero PC (2002) Surgical treatment of non-small cell Rusch VW, Parekh KR, Leon L et al (2000) Factors determin-
lung cancer 1 cm or less in diameter. Ann Thorac Surg ing outcome after surgical resection of T3 and T4 lung
73:1545–1550 (discussion 1550–1551) cancers of the superior sulcus. J Thorac Cardiovasc Surg
Morgan JA, Ginsburg ME, Sonett JR, Argenziano M, Walker 119:1147–1153
WS (2003) Thoracoscopic lobectomy using robotic technol- Rusch VW, Asamura H, Watanabe H, Giroux DJ, Rami-Porta
ogy video-assisted thoracic surgery: pulmonary lobectomy. R, Goldstraw P (2009) The IASLC lung cancer staging
Heart Surg Forum 6:233–244 project: a proposal for a new international lymph node map
Morikawa T, Katoh H, Takeuchi E, Ohbuchi T (1998) in the forthcoming seventh edition of the TNM classifica-
Technical feasibility of video-assisted lobectomy with tion for lung cancer. J Thorac Oncol 4:568–577
radical lymphadenectomy for primary lung cancer. Surg Schuchert MJ, Kilic A, Pennathur A et al (2011) Oncologic
Laparosc Endosc 8:466–473 outcomes after surgical resection of subcentimeter non-
Mountain CF, Dresler CM (1997) Regional lymph node small cell lung cancer. Ann Thorac Surg 91:1681–1688
classification for lung cancer staging. Chest 111:1718–1723 Shaw RR, Paulson DL, Kee JL (1961) Treatment of the
Myrdal G, Gustafsson G, Lambe M, Horte LG, Stahle E (2001) superior sulcus tumor by irradiation followed by resection.
Outcome after lung cancer surgery. Factors predicting early Ann Surg 154:29–40
mortality and major morbidity. Eur J Cardiothorac Surg Shim SS, Lee KS, Kim BT et al (2005) Non-small cell lung
20:694–699 cancer: prospective comparison of integrated FDG PET/CT
Naruke T, Suemasu K, Ishikawa S (1978) Lymph node and CT alone for preoperative staging. Radiology
mapping and curability at various levels of metastasis in 236:1011–1019
resected lung cancer. J Thorac Cardiovasc Surg 76:832–839 Shiotani S, Sugimura K, Sugihara M et al (2000) Diagnosis of
National Cancer Institute (2010) Phase III randomized study of chest wall invasion by lung cancer: useful criteria for
lobectomy versus sublobar resection in patients with small exclusion of the possibility of chest wall invasion with MR
peripheral stage IA non-small cell lung cancer. In: NCI imaging. Radiat Med 18:283–290
clinical trials (PDQ) Silvestri GA, Tanoue LT, Margolis ML, Barker J, Detterbeck F,
Ohbuchi T, Morikawa T, Takeuchi E, Kato H (1998) Lobec- American College of Chest Physicians (2003) The nonin-
tomy: video-assisted thoracic surgery versus posterolateral vasive staging of non-small cell lung cancer: the guidelines.
thoracotomy. Jpn J Thorac Cardiovasc Surg 46:519–522 Chest 123:147S–156S
Oldenburg FA Jr, Newhouse MT (1979) Thoracoscopy. A safe, Stephan F, Boucheseiche S, Hollande J et al (2000) Pulmonary
accurate diagnostic procedure using the rigid thoracoscope complications following lung resection: a comprehensive
and local anesthesia. Chest 75:45–50 analysis of incidence and possible risk factors. Chest
Ost D, Fein AM, Feinsilver SH (2003) Clinical practice. The 118:1263–1270
solitary pulmonary nodule. N Engl J Med 348:2535–2542 Sugarbaker DJ, Swanson SJ, Shen RK (2001) Pulmonary
Park BJ, Flores R, Downey RJ, Bains MS, Rusch VW (2003) resection, 4th edn. Lippincott Williams and Wilkins,
Management of major hemorrhage during mediastinoscopy. Philadelphia
J Thorac Cardiovasc Surg 126:726–731 Sugi K, Kaneda Y, Esato K (2000) Video-assisted thoraco-
Patchell RA, Tibbs PA, Walsh JW et al (1990) A randomized scopic lobectomy achieves a satisfactory long-term prog-
trial of surgery in the treatment of single metastases to the nosis in patients with clinical stage IA lung cancer. World J
brain. N Engl J Med 322:494–500 Surg 24:27–30 (discussion -1)
Piehler JM, Pairolero PC, Weiland LH, Offord KP, Payne WS, Swanson SJ, Herndon JE, D’Amico TA et al (2007) Video-
Bernatz PE (1982) Bronchogenic carcinoma with chest wall assisted thoracic surgery lobectomy: report of CALGB
invasion: factors affecting survival following en bloc 39802—a prospective, multi-institution feasibility study.
resection. Ann Thorac Surg 34:684–691 J Clin Oncol 25:4993–4997
Lung Cancer Surgery 117

Takizawa T, Terashima M, Koike T et al (1998) Lymph node Walker WS, Codispoti M, Soon SY, Stamenkovic S, Carnochan
metastasis in small peripheral adenocarcinoma of the lung. F, Pugh G (2003) Long-term outcomes following VATS
J Thorac Cardiovasc Surg 116:276–280 lobectomy for non-small cell bronchogenic carcinoma. Eur
Tanaka K, Kubota K, Kodama T, Nagai K, Nishiwaki Y (1999) J Cardiothorac Surg 23:397–402
Extrathoracic staging is not necessary for non-small-cell Warren WH, Faber LP (1994) Segmentectomy versus lobect-
lung cancer with clinical stage T1-2 N0. Ann Thorac Surg omy in patients with stage I pulmonary carcinoma. Five-
68:1039–1042 year survival and patterns of intrathoracic recurrence.
Tatsumi A, Ueda Y (2003) Video-assisted thoracic surgery for J Thorac Cardiovasc Surg 107:1087–1093 (discussion
lung cancer: is it a feasible operation for stage I lung 1093–1094)
cancer? Jpn J Thorac Cardiovasc Surg 51:646–650 Webb WR, Gatsonis C, Zerhouni EA et al (1991) CT and MR
Toloza EM, Harpole L, McCrory DC (2003a) Noninvasive imaging in staging non-small cell bronchogenic carcinoma:
staging of non-small cell lung cancer: a review of the report of the Radiologic Diagnostic Oncology Group.
current evidence. Chest 123:137S–146S Radiology 178:705–713
Toloza EM, Harpole L, Detterbeck F, McCrory DC (2003b) Weng E, Tran L, Rege S et al (2000) Accuracy and clinical
Invasive staging of non-small cell lung cancer: a review of impact of mediastinal lymph node staging with FDG-PET
the current evidence. Chest 123:157S–166S imaging in potentially resectable lung cancer. Am J Clin
Tovar EA, Roethe RA, Weissig MD, Lloyd RE, Patel GR Oncol 23:47–52
(1998) One-day admission for lung lobectomy: an incidental Wernly JA, DeMeester TR, Kirchner PT, Myerowitz PD,
result of a clinical pathway. Ann Thorac Surg 65:803–806 Oxford DE, Golomb HM (1980) Clinical value of quanti-
Travis W, Brambilla E, Muller-Hermelink H, Harris C (eds) tative ventilation–perfusion lung scans in the surgical
(2004) World Health Organization classification of tumors. management of bronchogenic carcinoma. J Thorac Cardio-
IARC Press, Lyon vasc Surg 80:535–543
Travis WD, Brambilla E, Noguchi M et al (2011) International Wright CD, Wain JC, Grillo HC, Moncure AC, Macaluso SM,
association for the study of lung cancer/American Thoracic Mathisen DJ (1997) Pulmonary lobectomy patient care
Society/European Respiratory Society international multi- pathway: a model to control cost and maintain quality. Ann
disciplinary classification of lung adenocarcinoma. J Thorac Thorac Surg 64:299–302
Oncol 6:244–285 Wright CD, Menard MT, Wain JC et al (2002) Induction
Tschernko EM, Hofer S, Bieglmayer C, Wisser W, Haider W chemoradiation compared with induction radiation for lung
(1996) Early postoperative stress: video-assisted wedge cancer involving the superior sulcus. Ann Thorac Surg
resection/lobectomy vs conventional axillary thoracotomy. 73:1541–1544
Chest 109:1636–1642 Yano T, Yokoyama H, Fukuyama Y, Takai E, Mizutani K,
Van Schil PE (2007) Advantages in surgical staging. Thoraco- Ichinose Y (1997) The current status of postoperative
scopy or video-assisted thoracic surgery (VATS). J Thorac complications and risk factors after a pulmonary resection
Oncol 2:S224–S225 for primary lung cancer. A multivariate analysis. Eur J
Venissac N, Alifano M, Mouroux J (2003) Video-assisted Cardiothorac Surg 11:445–449
mediastinoscopy: experience from 240 consecutive cases. Yoshikawa K, Tsubota N, Kodama K, Ayabe H, Taki T, Mori T
Ann Thorac Surg 76:208–212 (2002) Prospective study of extended segmentectomy for
Walker WS, Carnochan FM, Tin M (1993) Thoracoscopy small lung tumors: the final report. Ann Thorac Surg
assisted pulmonary lobectomy. Thorax 48:921–924 73:1055–1058 (discussion 1058–1059)
Walker WS, Pugh GC, Craig SR, Carnochan FM (1996) Zielinski M, Rami-Porta R (2007) Proposals for changes in the
Continued experience with thoracoscopic major pulmonary Mountain and Dresler mediastinal and pulmonary lymph
resection. Int Surg 81:255–258 node map. J Thorac Oncol 2:3–6
Radiation Response of the Normal Lung
Tissue and Lung Tumors
Hiromitsu Iwata, Taro Murai, and Yuta Shibamoto

Contents Abstract
We reviewed recent investigations on radiation
1 Introduction.............................................................. 119 response of normal lung tissue and lung tumors
2 Radiation Response of Normal Lung Tissues ...... 120 and also introduced our own investigations. The
2.1 Mechanism of Radiation Injury ................................ 120 mechanisms for response of normal lung tissues to
2.2 Factors Related to Development of Radiation radiation are not yet fully understood. Recent
Pneumonitis................................................................ 121
researches have revealed that various cytokines
2.3 Cryptogenic Organizing Pneumonia After
Pulmonary Irradiation................................................ 122 and lung parenchymal cells are involved in the
pathogenesis of radiation response of normal lung
3 Radiation Response of Lung Tumors ................... 122
3.1 Prediction of Radiosensitivity and Proliferative tissues. Prediction of tumor and/or normal tissue
Activity of Tumors .................................................... 122 sensitivity to treatment is being investigated for
3.2 Tumor Progression During Waiting Time Before tailor-made cancer treatment based on the biolog-
Radiotherapy .............................................................. 124 ical characteristics. In the near future, it is hoped
3.3 Radiation Response by Tumor Histology................. 125
3.4 Haplotypes of Genes Associated with Risk that the relationship among clonogenic death of
of Adverse Normal Tissue Reactions ....................... 126 target cells, cytokine induction, gene expression
and radiation pneumonitis, and that among radio-
References.......................................................................... 127
sensitivity, tumor histology, tumor gene expression
and patient characteristics will be clarified. Future
investigations will lead to a step toward an era of
personalized cancer treatment.

1 Introduction

Radiation response of the normal lung tissue and lung


tumors has been studied for nearly 90 years. Pul-
monary damage complicating radiotherapy for breast
cancer was first reported in 1923 (Groover et al.
H. Iwata  T. Murai  Y. Shibamoto (&) 1923). Pathological changes in lung tissue following
Department of Radiology, chest wall irradiation have been subsequently repor-
Nagoya City University Graduate School of Medical
ted in the 1930–1950s (Engelstad 1940; Bergmann
Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku,
467-8601 Nagoya, Japan and Graham 1951). Radiotherapy for lung tumors
e-mail: yshiba@med.nagoya-cu.ac.jp moved into full swing in the early 1950s. Since then,

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 119
DOI: 10.1007/174_2011_250, Ó Springer-Verlag Berlin Heidelberg 2011
120 H. Iwata et al.

the effect of radiotherapy for lung tumors and lung injury caused by irradiation, for example, the type II
function after radiotherapy were reported one after pneumocyte has been considered to be one of the
another. (Sweany et al. 1959; Barton et al. 1960). most important target cells. The type II pneumocyte
During these times, Puck and Marcus (1956) devised exhibited the earliest response to radiation and a
a colony formation method with which survival of decrease in lamellar bodies and a corresponding
cells became measurable in vitro. This facilitated increase in alveolar surfactant were reported shortly
tremendous progress in radiation biology at the cel- after radiation (Penney et al. 1982). However, Rubin
lular levels. Most of the important biological phe- et al. (1995) demonstrated radiation-induced damage
nomena at the cellular and animal levels appeared to progression is the result of an early activation of an
have been clarified by 1980. Thereafter, advances in inflammatory reaction leading to the expression and
molecular biology have brought us much new maintenance of an elevated cytokine cascade and
knowledge on radiobiology of various normal tissues gene expression following pulmonary irradiation in
and tumors. Although molecular mechanisms for mice. Among these cytokines and genes are inter-
normal lung tissue reaction and lung tumor cell killing leukin 1 (IL-1), IL-6, transforming growth factor
are not completely clarified yet, many cytokines have beta-1 (TGF-b1), tumor necrosis factor alpha
been identified that are involved in the pathogenesis (TNF-a), high-molecular-weight mucin-like antigen
of normal lung tissue reaction including radiation KL-6, platelet-derived growth factor, and collagen
pneumonitis. Some of more recent researches aim at and fibronection genes. The pathogenesis of radiation
identifying single nucleotide polymorphism of genes, lung damage is considered to be similar to that of
oncogene expression and angiogenesis that are pos- interstitial pneumonitis. It has also been shown that
sibly related to increased normal lung tissue reaction inducible nitric oxide synthase (NOS) and nitric oxide
and radiosensitivity of lung tumors. are involved in radiation pneumonitis. Inducible NOS
In recent years, the use of stereotactic body is strongly expressed in pneumocytes and alveolar
radiotherapy (SBRT) has spread as a new treatment macrophages in idiopathic interstitial pneumonitis.
modality for lung tumors. Along with the develop- Peroxynitrite, a potent oxidant produced by the rapid
ment of treatment methods and machines, it has reaction of nitric oxide and superoxide, is highly
become necessary to evaluate radiation response of present in such lungs (Worthington et al. 2000; Saleh
normal lung tissue and lung tumors, especially to et al. 1997). Recent data have also shown that nitric
hypofractionated high-dose-per-fraction radiation oxide induces stabilization of hypoxia inducible fac-
delivery. In this article, we review recent investiga- tor 1, alpha (HIF-1a) through a mechanism involving
tions regarding these issues and also introduce our free radicals. Therefore, upregulation of NOS by
own investigations on radiation response of normal radiation may subsequently trigger the HIF/vascular
lung tissue and lung tumors. endothelial growth factor (VEGF) molecular cascade
(Quintero et al. 2006). As for chronic lung injury, Fas/
CD95 and Fas-ligand activation is associated with a
fibrotic response. Fas-ligand protein is present in
2 Radiation Response of Normal infiltrating lymphocytes and granulocytes, and
Lung Tissues expression of Fas is upregulated in alveolar and
bronchiolar epithelial cells (Kuwano et al. 1999).
2.1 Mechanism of Radiation Injury Radiation directly induces Fas expression in cells, and
inflammatory cells consecutively expressing Fas-
Although the mechanisms for response of normal lung ligand may promote apoptosis of pneumocytes. It has
tissues to radiation are not yet fully understood, recent also been shown that activated fibroblasts in the
researches have revealed that many factors and vari- induction of apoptosis of alveolar epithelial cells may
ous lung parenchymal cells contribute to the patho- play a role of chronic lung injury (Martin et al. 2005).
genesis of radiation response of normal lung tissues Therefore, if early apoptosis responses in the alveolar
(Kong et al. 2005). Previously, clonogenic death of cells are mediated by excess concentrations of Fas-
target cells has been considered to be a major cause of ligand, then blocking apoptosis early after lung injury
normal tissue injury by irradiation. Regarding lung might be beneficial.
Radiation Response of the Normal Lung Tissue and Lung Tumors 121

Table 1 Multivariate analysis of factors associated with the occurrence of C Grade 2 radiation pneumonitis
p Hazard ratio, p Hazard ratio,
95% CI 95% CI
KL-6 (U/ml) 0.04 6.1 (1.1–34.7) 0.02 6.8 (1.5–31.5)
(C500, \500)
Sex 0.10 6.1 (0.7–50.8) 0.10 6.0 (0.7–50.9)
(M, F)
PTV volume (cm3) 0.15 2.6 (0.7–9.1) 0.22 2.2 (0.6–7.5)
(C43, \43)a
Presence of IP 0.56 2.0 (0.2–22.6)
(+, -)
b
V20 Lung (%) 0.64 0.7 (0.1–3.8)
(C6, \6)a
Mean lung dose (Gy) 0.38 2.1 (0.4–11.6)
(C4, \4)a
Differences were tested by logistic regression
PTV planning target volume, IP interstitial pneumonitis, CI confidence interval
a
Divided by the median
b
Percentage of the lung receiving a 20 Gy or higher dose

2.2 Factors Related to Development management of severe radiation pneumonitis after


of Radiation Pneumonitis SBRT. The sialylated carbohydrate antigen KL-6 is a
circulating mucin-like glycoprotein with a high
Factors such as age, gender, smoking status, location molecular weight that is classified as a cluster 9
of tumor, and lung dosimetric factors can also con- human MUC-1 antigen. It has been found to be highly
tribute to the lung damage caused by radiation. Pre- expressed on type-2 pneumocytes and bronchiolar
vious reports have shown a correlation between epithelial cells (Kohno et al. 1988). MUC-1 is a large
severe radiation pneumonitis and dose-volume transmembrane glycoprotein that has a rigid structure
parameters such as the mean lung dose, V20 (per- protruding 200–500 nm above the plasma membrane
centage of the lung volume receiving C20 Gy), and and is found at the surface of normal glandular epi-
V5 (Tucker et al. 2010; Jin et al. 2009). Yet, it seems thelial cells (Stahel et al. 1994; Hilkens et al. 1992).
that factors other than these physical parameters also The serum levels of KL-6 have been shown to cor-
influence the incidence and severity of radiation relate with interstitial pneumonia rather than with
pneumonitis. With respect to conventional radiother- other benign diseases of the lung and other organs
apy cases, recent researchers have revealed that (Kohno et al. 1989; Kobayashi and Kitamura 1995).
appropriate inhibition of key cytokines at an early Moreover, several reports have shown that KL-6 is
stage might provide new tools for the effective treat- useful for early diagnosis of severe radiation pneu-
ment of radiation pneumonitis (Anscher et al. 2003; monitis in patients undergoing conventional thoracic
Goto et al. 2001). However, in patients undergoing radiotherapy (Goto et al. 2001; Matsuno et al. 2006).
SBRT for lung tumors, only a few reports have sug- However, in patients undergoing SBRT for lung
gested that the early response of blood markers tumors, only a few reports have suggested that KL-6
mentioned above is a good indicator of severe radia- is a good indicator of severe radiation pneumonitis
tion pneumonitis. (Hara et al. 2004; Yamashita et al. 2010).
In February 2004, we started SBRT for stage I Table 1 shows the results of multivariate analysis
NSCLC and lung metastasis with our own protocols in our patients undergoing SBRT for stage I lung
(Baba et al. 2010). During the protocol-based study, cancer or lung metastasis. First, six factors that
we have investigated the usefulness of KL-6 mea- showed a p value of \0.2 in the univariate analysis
surement for predicting the occurrence and were analyzed, and only the KL-6 level proved to be a
122 H. Iwata et al.

2500 is high, lowering the prescribed dose or avoiding


SBRT may be difficult. In such patients, therefore,
pretreatment with blood markers should be used to
2000
predict the occurrence of radiation pneumonitis (Iwata
et al., submitted for publication).

1500 2.3 Cryptogenic Organizing Pneumonia


KL–6 (U/ml)

After Pulmonary Irradiation

1000 Recently, unusual pneumonitis after radiotherapy of


breast-conserving therapy was reported (Ogo et al.
*
2008; Katayama et al. 2009; Takigawa et al. 2000;
Kubo et al. 2009). It is characterized by lung infiltrates
500 *
outside the radiation field, which are different from
*
radiation pneumonitis. The clinical features resemble
cryptogenic organizing pneumonia (COP). Radiation
0 pneumonitis usually occurs within several months
–3 –2 –1 0 1 2 3 4 5 after completion of radiotherapy and is generally
Months
limited to the irradiated field; furthermore, migration
Fig. 1 Serum KL-6 levels (cutoff level: 500 U/ml) of patients of shadows is not observed in radiation pneumonitis.
who developed Grade C2 radiation pneumonitis. Month 0 The incidence was reported to be 1.8–2.9%. We
represents the month when radiation pneumonitis first devel- investigated clinical and radiological features of
oped. The asterisks indicate the occurrence of cryptogenic
unusual pneumonitis like COP occurring after SBRT
organizing pneumonia after a decrease in the KL-6 level in
response to steroid administration of the lung and its incidence (Murai et al. 2010). This
was the first cohort study after SBRT. The incidence
significant factor related to the occurrence of [ Grade of unusual pneumonitis like COP after SBRT was
2 radiation pneumonitis. Then, three factors (pre- 4.9%, and appeared to be somewhat higher than that
treatment serum KL-6 level, gender, and PTV vol- reported for patients undergoing tangential breast
ume) that showed a p value of \0.05 were analyzed, irradiation (Table 2). The symptoms were sometimes
and again, only the KL-6 level proved to be signifi- severe, but steroids were effective. These patients with
cant. In our study, KL-6 levels increased almost in unusual pneumonitis like COP had a history of pre-
accordance with the occurrence of C Grade 2 radia- vious symptomatic radiation pneumonitis before
tion pneumonitis and decreased in accordance with the COP-like pneumonitis. Figure 2 shows the example of
response to steroid administration (Fig. 1). Moreover, unusual pneumonitis like COP after SBRT for pri-
at the onset of Grade 2 or 3 radiation pneumonitis, mary lung cancer. We could not find obvious risk
KL-6 levels were increased by 1.7 times on average factors. Further investigation appears to be warranted
compared to the pre-treatment level. We think that to identify its pathogenesis and risk factor.
serial measurement of serum KL-6 levels before and
after SBRT would help to predict the occurrence of
radiation pneumonitis and manage severe radiation 3 Radiation Response of Lung
pneumonitis after SBRT. If it is proven to be accurate Tumors
predictors for severe radiation pneumonitis, the blood
markers and the models of recommended dose volume 3.1 Prediction of Radiosensitivity
limits for predicting radiation pneumonitis may permit and Proliferative Activity of Tumors
us to select low-risk patients for dose escalation to
improve tumor local control and overall survival The necessity for tailor-made cancer treatment based
without increasing radiation damage. On the other on the biological characteristics of each tumor has
hand, even if the risk of severe radiation pneumonitis been advocated since three decades ago. For this
Radiation Response of the Normal Lung Tissue and Lung Tumors 123

Table 2 Representative reported results of unusual pneumonitis like cryptogenic organizing pneumonia occurring after
radiotherapy
Author Treatment N Occurrence frequency (%) Relative factor
Ogo (2008) BCT 2056 1.8 None
Katayama (2009) BCT 702 2.3 Age, tamoxifen
Takigawa (2000) BCT 157 2.5 None
Kubo (2009) BCT 413 2.9 Central lung distance
Total BCT 3328 2.1*
Murai (2010) SBRT 164 4.9* Planning target volume
BCT breast-conserving therapy, SBRT stereotactic body radiotherapy
*
There was a significant difference between representative reported results after BCT and our data after SBRT (p = 0.02, Chi-
square test)

Fig. 2 Chest CT, radiograph and dose distribution of stereo- Unusual pneumonitis like cryptogenic organizing pneumonia
tactic body radiotherapy (SBRT) for T2N0M0 primary lung occurred 3 months after resolution of radiation pneumonitis.
cancer of a 82-year-old patient. He presented with radiation Areas receiving more than 0.5 Gy are bounded by white lines
pneumonitis around planning target volume with Grade III (e and f). Chest radiograph shows newly developed infiltrates in
dyspnea at 8 months after SBRT (a and b, yellow arrow). the right lung outside the volume of [0.5 Gy (c and d, red
Radiation pneumonitis was resolved using prednisolone. arrow)

purpose, prediction of tumor and/or normal tissue cells at 2 Gy of in vitro irradiation is measured using
sensitivity to treatment is necessary. To estimate colony formation or colorimetric methods was most
radiosensitivity of tumor cells, several types of pre- intensively investigated (West et al. 1997). However,
dictive assays have been proposed. Among them, the the use of the SF-2 assay for radiosensitivity predic-
SF-2 assay in which the surviving fraction of tumor tion did not become a commonly used tool in clinics.
124 H. Iwata et al.

Fig. 3 Rate of increase in


volume and waiting time in Adenocarcinoma Squamous cell carcinoma
lung adenocarcinoma and
400
squamous cell carcinoma.
Both subtypes showed a
correlation between the two;
that is, the rate of increase
became higher with p = 0.02 p < 0.001
elongation of waiting time. 300

Volume increase rate (%)


Modified from Murai et al.
(2011)

200

100

0 8 16 24 32 0 8 16 24 32

Waiting time (weeks)

The reasons for this may include labor intensiveness proliferative activity and radiosensitivity according to
and a long waiting time before obtaining assay histological type of tumor were not clarified.
results. Shibamoto et al. (1994, 1998) tried to estab-
lish a more rapid assay of radiosensitivity using the
cytokinesis-block micronucleus (MN) test. MN for- 3.2 Tumor Progression During Waiting
mation represents chromosomal damage and the MN Time Before Radiotherapy
frequency increases with radiation dose. Using this
assay, a method of simultaneously estimating radio- An important issue to be addressed in clinical radio-
sensitivity and proliferative activity of human tumors therapy is the influence of waiting time (WT) before
was devised (Shibamoto et al. 1994, 1998). Estima- cancer treatment. The use of SBRT for stage I lung
tion of tumor proliferative activity is also important in cancer is rapidly increasing, so long WTs seem to
radiotherapy, since rapidly-growing tumors are con- have become a problem in many institutions. In head-
sidered to be resistant to protracted conventional and-neck cancer, long WTs are known to adversely
radiotherapy. One of the important parameters of affect treatment outcome; in a meta-analysis by Chen
proliferative activity is the potential doubling time et al. (2008), the relative risk of local recurrence was
(Tpot). The Tpot which is a doubling time in the estimated to increase by 1.15 times if patients were
absence of the cell loss is considered to represent required to wait for a month or longer before the
repopulation rates during and after radiotherapy better initiation of definitive radiotherapy. Murai et al.
than the volume doubling time. The Tpot varies (2011) investigated the relationship between WT and
greatly according to the histology of each tumor. disease progression in patients undergoing SBRT for
Radiosensitivity and proliferative activity character- lung adenocarcinoma (AD) or squamous cell carci-
istics of primary lung cancers determined using this noma (SQ). The median WT was 42 days (range,
assay were published previously (Shibamoto et al. 5–323 days). Figure 3 shows the correlation between
1998). The data showed that a high proliferative WT and rate of increase in volume in both AD and
activity was associated with an increased recurrence SQ. The median volume doubling time of AD and SQ
rate after operation. However, differences in was 170 and 93 days, respectively. Thirty-six tumors
Radiation Response of the Normal Lung Tissue and Lung Tumors 125

(23%) did not show volume increase during Adenocarcinoma (n=32)


WTs [ 25 days. In 41 patients waiting for B4 weeks, Squamous cell carcinoma (n=23)
no patient showed T-stage progression, whereas in 25
of 120 (21%) patients waiting for [4 weeks, T stage 100
progressed from T1 to T2 (p = 0.001). In 10 of 110
(9.1%) T1 ADs and 15 of 51 (29%) T1 SQs, T stage 80

Overall survival(%)
progressed (p = 0.002). The risk of T-stage progres-
sion in T1 lung cancer was significantly higher in the 60

group waiting for longer than 4 weeks. Therefore, this


40
study supports the recommendation of the British
Thoracic Society (1998) that the WT should be less
20
than 4 weeks in patients undergoing curative surgery.
In addition, 4 weeks may be appropriate for accept-
0
able WT before SBRT, although the WT should be as
short as possible. 0 12 24 36 48 60 72

100

3.3 Radiation Response by Tumor 80


Histology Local control (%)
60
Radiation sensitivity is known to differ with tumor
histology. In primary lung cancer, small cell carci- 40
nomas are generally considered to be more radio-
sensitive than adenocarcinomas and squamous cell 20

carciniomas, but this impression may be largely due


0
to the rapid response of small cell carcinomas to
radiation. Ultimate local control rates might not be so 0 12 24 36 48 60 72
different among the three histological subtypes of
100
lung cancer (Ruckdeschel 1998; Brodin et al. 1991).
Disease-free survival (%)

During carbon-ion radiotherapy performed at the 80


National Institute of Radiological Sciences, Chiba,
squamous cell carcinomas tended to show lower local 60
control rates than melanomas and adenoid cystic
carcinomas (Mizoe et al. 2004). During proton ther- 40
apy and carbon-ion radiotherapy performed for head-
and-neck cancer at Hyogo Ion Beam Medical Center, 20
a similar trend was also experienced (Murakami et al.
2009). A similar trend was also observed for particle 0
therapy of primary lung cancer. We evaluated the 0 12 24 36 48 60 72
clinical outcome of particle therapy for stage I non- Time (months)
small-cell lung cancer (Iwata et al. 2010). Figure 4
shows overall survival, local control and disease-free Fig. 4 Overall survival, local control, and disease-free sur-
vival curves for patients treated with proton therapy according
survival curves according to histology in patients
to histology. Patients with adenocarcinoma had a higher local
treated with proton therapy. The local control rates control rate than those with squamous cell carcinoma
were higher for adenocarcinoma than for squa- (p = 0.022), although the overall and disease-free survival
mous cell carcinoma (p = 0.02). This is in contrast to rates were not different (p = 0.19 and 0.061, respectively).
Modified from Iwata et al. (2010)
the laboratory and clinical observations for
126 H. Iwata et al.

radiosensitivity of NSCLC cells to X-rays (Shibamoto previous systemic chemotherapy or local therapy in
et al. 1998; Koukourakis et al. 1996). The differences future studies.
in radiosensitivity shown by histology should also be In the near future, it is hoped that the relationship
investigated further, and more studies are necessary to among clonogenic death of target cells, cytokine
prove whether these observations are correct. At induction, gene expression, and radiation pneumoni-
present, it may be concluded that particle radio- tis, and that among radiosensitivity for each tumor
therapy is efficacious against adenocarcinoma, which histology, tumor gene expression and patient charac-
may not be well responsive to conventional teristics will be clarified. Future discoveries will lead
radiotherapy. a step toward an era of personalized cancer treatment.
With respect to radiotherapy for lung metastasis,
large-scale data on outcome according to the primary
site or histology have not been reported to the best of 3.4 Haplotypes of Genes Associated
our knowledge. Milano et al. (2008) reported that with Risk of Adverse Normal Tissue
patients with oligometastatic disease in the lung, Reactions
brain, or liver etc. from primary breast cancer had a
significantly better outcome than those with other The RadGenomics project started in April 2001 at the
primary tumors. On the other hand, it was reported National Institute of Radiological Sciences in Japan
that oligometastases confined to one organ from (Iwakawa et al. 2002). This project promotes analysis
colorectal cancer treated by SBRT showed a 3-year of genes that are expressed in response to irradiation,
local control of 66% and a 5-year local control rate of identification of their allelic variants in the human
only 24% (Kang et al. 2010). Also, recent data have population, development of an effective procedure for
shown that 3-year local control rate after SBRT using quantitating individual radiosensitivity, and analysis
3 fractions for isolated colorectal lung metastases was of the relationship between genetic heterogeneity and
as low as 53% (Kim et al. 2009). Although the total susceptibility to irradiation. Major groups of genes
dose is related to local control rates (Kang et al. investigated in the project include DNA repair genes,
2010), a recent phase II study on lung metastasis from genes for programmed cell death, genes for signal
variable primary cancers treated by SBRT revealed a transduction, and genes for oxidative processes.
2-year local control rate of greater than 90% In a recent report, the global haplotype association
(Rusthoven et al. 2009). Herfarth et al. (2001) analysis (p \ 0.05 and false discovery rate \0.05)
observed that poorer control is achieved by SBRT for indicated that estimated haplotypes in six loci were
liver metastasis of colorectal cancer than for metas- associated with the risk of early adverse skin reactions
tases with other histology (45 vs. 91%, respectively, (EASR) with breast cancer (Suga et al. 2007). In the
at 18 months). There were many reports with radio- CD44 gene, the haplotype GGTT significantly
therapy for brain metastasis according to histology. increased the risk of EASRs compared with the most
Brain metastases of renal cell carcinoma, colorectal common haplotype GGTC (odds ratio = 2.17; 95%
cancer, and melanoma etc. were reported as radiore- CI, 1.07–4.43). Five haplotypes, CG in MAD2L2,
sistant (Meyners et al. 2010; Wronski et al. 1997; GTTG in PTTG1, TCC and CCG in RAD9A, and
Kruser et al. 2008). With respect to our data, lung GCT in LIG3 were associated with a reduced EASR
metastases from variable primary cancers treated by risk. Results of similar analyses in patients with
SBRT showed a 3-year local control of 88% in 70 uterine cervical cancer have recently published; two
lesions. In this study, the 3-year local control rate was haplotypes were associated with an increased risk of
lower for colorectal cancer metastases than for early adverse reaction in the gastrointestinal tract
metastases from other primary tumors (57 vs. 93%, (Ishikawa et al. 2011). Cervical cancer patients who
p = 0.0052; unpublished data). Thus, metastases have both risk haplotypes of NPAT-ATM and AU-
from colorectal cancer seem to be generally more RKA showed significant association with an increased
radioresistant than those from other primary tumors. risk of an early intestinal reaction to radiation therapy
However, since tumor cells could develop resistance (odds ratio = 3.24; 95% CI, 1.52–6.92).
following antitumor treatment failure, it is necessary Correlation between pulmonary toxicities and
to evaluate the radiation sensitivity in relation to single nucleotide polymorphisms of genes is also
Radiation Response of the Normal Lung Tissue and Lung Tumors 127

being investigated in the RadGenomic project, along Iwakawa M, Imai T, Harada Y et al (2002) RadGenomic
with the breast cancer and uterine cervical cancer project. J Jpn Radiol Soc 62:484–489
Iwata H, Murakami M, Demizu Y et al (2010) High-dose
studies. Radiation pneumonitis is one of the major proton therapy and carbon-ion therapy for stage I nonsmall
normal tissue reactions investigated in this collabo- cell lung cancer. Cancer 116:2476–2485
ration. Analyses are ongoing and the results will be Jin H, Tucker SL, Liu HH et al (2009) Dose-volume thresholds
obtained in the near future. and smoking status for the risk of treatment-related
pneumonitis in inoperable non-small cell lung cancer
treated with definitive radiotherapy. Radiother Oncol
91:427–432
References Kang JK, Kim MS, Kim JH et al (2010) Oligometastases
confined one organ from colorectal cancer treated by SBRT.
Clin Exp Metastasis 27:273–278
Anscher MS, Marks LB, Shafman TD et al (2003) Risk of long- Katayama N, Sato S, Katsui K et al (2009) Analysis of factors
term complications after TFG-beta1-guided very-high-dose associated with radiation-induced bronchiolitis obliterans
thoracic radiotherapy. Int J Radiat Oncol Biol Phys 56:988– organizing pneumonia syndrome after breast-conserving
995 therapy. Int J Radiat Oncol Biol Phys 73:1049–1054
Baba F, Shibamoto Y, Ogino H et al (2010) Clinical outcomes Kim MS, Yoo SY, Cho CK et al (2009) Stereotactic body
of stereotactic body radiotherapy for stage I non-small cell radiation therapy using three fractions for isolated lung
lung cancer using different doses depending on tumor size. recurrence from colorectal cancer. Oncology 76:212–219
Radiat Oncol 5:81 Kobayashi J, Kitamura S (1995) KL-6: a serum marker for
Barton HL, Mcgrahan GM Jr, Jordan GL Jr (1960) The interstitial pneumonia. Chest 108:311–315
evaluation of roentgen therapy in the management of non- Kohno N, Akiyama M, Kyoizumi S et al (1988) Detection of
resectable carcinoma of the lung. Dis Chest 37:170–175 soluble tumor-associated antigens in sera and effusions
Bergmann M, Graham EA (1951) Pneumonectomy for severe using novel monoclonal antibodies, KL-3 and KL-6, against
irradiation damage of the lung. J Thorac Surg 22:549–567 lung adenocarcinoma. Jpn J Clin Oncol 18:203–216
British Thoracic Society (1998) BTS recommendations to Kohno N, Kyoizumi S, Awaya Y et al (1989) New serum
respiratory physicians for organising the care of patients indicator of interstitial pneumonitis activity. Sialylated
with lung cancer. The lung cancer working party of the carbohydrate antigen KL-6. Chest 96:68–73
British thoracic society standards of care committee. Thorax Kong FM, Ten Haken R, Eisbruch A, Lawrence TS (2005)
53:S1–S8 Non-small cell lung cancer therapy-related pulmonary
Brodin O, Lennartsson L, Nilsson S (1991) Single-dose and toxicity: an update on radiation pneumonitis and fibrosis.
fractionated irradiation of four human lung cancer cell lines Semin Oncol 32:S42–S54
in vitro. Acta Oncol 30:967–974 Koukourakis M, Hlouverakis G, Kosma L et al (1996) The
Chen Z, King W, Pearcey R, Kerba M, Mackillop WJ (2008) impact of overall treatment time on the results of radio-
The relationship between waiting time for radiotherapy and therapy for non-small cell lung carcinoma. Int J Radiat
clinical outcomes: a systematic review of the literature. Oncol Biol Phys 34:315–322
Radiother Oncol 87:3–16 Kruser TJ, Chao ST, Elson P et al (2008) Multidisciplinary
Engelstad RB (1940) Pulmonary lesions after roentgen and management of colorectal brain metastases. A retrospective
radium irradiation. Am J Roentgenol 43:676 study. Cancer 113:158–165
Goto K, Kodama T, Sekine I et al (2001) Serum levels of KL-6 Kubo A, Osaki K, Kawanaka T et al (2009) Risk factors for
are useful biomarkers for severe radiation pneumonitis. radiation pneumonitis caused by whole breast irradiation
Lung Cancer 34:141–148 following breast-conserving surgery. J Med Invest 56:99–
Groover TA, Christie AC, Merritt EA (1923) Intrathoracic 110
changes following roentgen treatment of breast carcinoma. Kuwano K, Miyazaki H, Hagimoto N et al (1999) The
Am J Roentgenol 10:471 involvement of Fas-Fas ligand pathway in fibrosing lung
Hara R, Itami J, Komiyama T et al (2004) Serum levels of KL-6 diseases. Am J Respir Cell Mol Biol 20:53–60
for predicting the occurrence of radiation pneumonitis after Martin TR, Hagimoto N, Nakamura M, Matute-Bello G (2005)
stereotactic radiotherapy for lung tumors. Chest 125:340– Apoptosis and epithelial injury in the lungs. Proc Am
344 Thorac Soc 2:214–220
Herfarth KK, Debus J, Lohr F et al (2001) Stereotactic single- Matsuno Y, Satoh H, Ishikawa H et al (2006) Simultaneous
dose radiation therapy of liver tumors: results of a phase I/II measurements of KL-6 and SP-D in patients undergoing
trial. J Clin Oncol 19:164–170 thoracic radiotherapy. Med Oncol 23:75–82
Hilkens J, Ligtenberg MJ, Vos HL et al (1992) Cell membrane- Meyners T, Heisterkamp C, Kueter JD et al (2010) Prognostic
associated mucins and their adhesion-modulating property. factors for outcomes after whole-brain irradiation of brain
Trends Biochem Sci 17:359–363 metastases from relatively radioresistant tumors: a retro-
Ishikawa A, Suga T, Shoji Y et al (2011) Genetic variants of spective analysis. BMC Cancer 10:582
NPAT-ATM and AURKA are associated with an early Milano MT, Katz AW, Muhs AG et al (2008) A prospective
adverse reaction in cervical cancer patients after radiation pilot study of curative-intent stereotactic body radiation
therapy. Int J Radiat Oncol Biol Phys (In press, 2010 Nov 2, therapy in patients with 5 or fewer oligometastatic lesions.
Epub ahead of print) Cancer 112:650–658
128 H. Iwata et al.

Mizoe JE, Tsujii H, Kamada T et al (2004) Dose escalation Shibamoto Y, Shibata T, Miyatake S et al (1994) Assessment of
study of carbon ion radiotherapy for locally advanced head- the proliferative activity and radiosensitivity of human
and-neck cancer. Int J Radiat Oncol Biol Phys 60:358– tumours using the cytokinesis-block micronucleus assay. Br
364 J Cancer 70:67–71
Murai T, Shibamoto Y, Baba F et al (2010) Cryptogenic Shibamoto Y, Ike O, Mizuno H, Fukuse T, Hitomi H,
organizing pneumonia after stereotactic body radiotherapy Takahashi M (1998) Proliferative activity and micronucleus
of the lung. Int J Radiat Oncol Biol Phys 78:S530–S531 frequency after radiation of lung cancer cells as assessed by
Murai T, Shibamoto Y, Baba F et al (2011) Progression of non- the cytokinesis-block method and their relationship to
small-cell lung cancer during the interval before stereotactic clinical outcome. Clin Cancer Res 4:677–682
body radiotherapy. Int J Radiat Oncol Biol Phys (In press, Stahel RA, Gilks WR, Lehmann HP et al (1994) Third
2010 Nov 20, Epub ahead of print) international workshop on lung tumor and differentiation
Murakami M, Demizu Y, Niwa Y et al (2009) Current status of antigens: overview of the results of the central data analysis.
the HIBMC, providing particle beam radiation therapy for Int J Cancer 8:S6–S26
more than 2,600 patients, and the prospects of laser-driven Suga T, Ishikawa A, Kohda M et al (2007) Halotype-based
proton radiotherapy. GermanyMunich, World congress analysis of genes associated with risk of adverse skin
2009, Medical physics and biomedical engineering (WC reactions after radiotherapy in breast cancer patients. Int J
2009) IFMBE proceedings, vol 25/1, pp 878–882. doi: Radiat Oncol Biol Phys 69:685–693
10.1007/978-3-642-03474-9_247 Sweany SK, Moss WT, Haddy FJ (1959) The effects of chest
Ogo E, Komaki R, Fujimoto K et al (2008) A survey of irradiation of pulmonary function. J Clin Invest 38:587–593
radiation-induced bronchiolitis obliterans organizing pneu- Takigawa N, Segawa Y, Saeki T et al (2000) Bronchiolitis
monia syndrome after breast-conserving therapy in Japan. obliterans organizing pneumonia syndrome in breast-con-
Int J Radiat Oncol Biol Phys 71:123–131 serving therapy for early breast cancer: radiation-induced
Penney DP, Siemann DW, Rubin P, Shapiro DL, Finkelstein J, lung toxicity. Int J Radiat Oncol Biol Phys 48:751–755
Cooper RA Jr (1982) Morphologic changes reflecting early Tucker SL, Jin H, Wei X et al (2010) Impact of toxicity grade
and late effects of irradiation of the distal lung of the mouse: and scoring system on the relationship between mean lung
a review. Scan Electron Microsc Pt 1:413–425 dose and risk of radiation pneumonitis in a large cohort of
Puck TT, Marcus PI (1956) Action of X-rays on mammalian patients with non-small cell lung cancer. Int J Radiat Oncol
cells. J Exp Med 103:653–666 Biol Phys 77:691–698
Quintero M, Brennan PA, Thomas GJ, Moncada S (2006) Nitric West CML, Davidson SE, Roberts SA, Hunter RD (1997) The
oxide is a factor in the stabilization of hypoxia-inducible independence of intrinsic radiosensitivity as a prognostic
factor-1alpha in cancer: role of free radical formation. factor for patient response to radiotherapy of carcinoma of
Cancer Res 66:770–774 the cervix. Br J Cancer 76:1184–1190
Rubin P, Johnson CJ, Williams JP, McDonald S, Finkelstein JN Worthington J, Robson T, Murray M, O’Rourke M, Keilty G,
(1995) A perpetual cascade of cytokines postirradiation Hirst DG (2000) Modification of vascular tone using iNOS
leads to pulmonary fibrosis. Int J Radiat Oncol Biol Phys under the control of a radiation-inducible promoter. Gene
33:99–109 Ther 7:1126–1131
Ruckdeschel JC (1998) Future directions in non-small-cell lung Wronski M, Maor MH, Davis BJ et al (1997) External radiation
cancer: a continuing perspective. Oncology 12:S90–S96 of brain metastases from renal carcinoma: a retrospective
Rusthoven KE, Kavanagh BD, Burri SH et al (2009) Multiin- study of 119 patients from the M.D. Anderson Cancer
stitutional phase I/II trial of stereotactic body radiation Center. Int J Radiat Oncol Biol Phys 37:753–759
therapy for lung metastases. J Clin Oncol 27:1579–1584 Yamashita H, Kobayashi-Shibata S, Terahara A et al (2010)
Saleh D, Barnes PJ, Giaid A (1997) Increased production of the Prescreening based on the presence of CT-scan abnormal-
potent oxidant peroxynitrite in the lungs of patients with ities and biomarkers (KL-6 and SP-D) may reduce severe
idiopathic pulmonary fibrosis. Am J Respir Crit Care Med radiation pneumonitis after stereotactic radiotherapy. Radiat
155:1763–1769 Oncol 5:32
Radiation Time, Dose, and Fractionation
in the Treatment of Lung Cancer
Daniel Gomez, Melenda D. Jeter, Ritsuko Komaki,
and James D. Cox

Contents Abstract
Radiation therapy has been an important component
1 Non-Small Cell Lung Cancer................................. 130 of potentially curative treatment of lung cancer for
1.1 Dose Escalation with Standard Fractionation .......... 130 more than 40 years. The radiosensitivity of normal
1.2 Large-Dose Fractionation and Stereotactic Body
Radiation Therapy ..................................................... 131
tissues in the thorax, especially the normal lung and
1.3 Dose Escalation with Hyperfractionation ................. 132 esophagus, has led to efforts to enhance the antitumor
1.4 Accelerated Fractionation.......................................... 132 effects of radiation while reducing its acute and late
1.5 Reducing the Target Volume with 3D Conformal adverse effects on normal tissues. Improving local
Radiation Therapy ..................................................... 133
1.6 Intensity-Modulated Radiation Therapy ................... 133
control of medically inoperable or locally advanced
1.7 Accounting for Tumor Motion ................................. 134 unresectable disease (stage III non-small cell lung
1.8 Proton Therapy .......................................................... 134 cancer or limited-stage small cell lung cancer, defined
1.9 Concurrent Chemotherapy......................................... 135 as disease confined to one hemithorax and the
2 Small Cell Lung Cancer ......................................... 135 ipsilateral supraclavicular lymph nodes) can favor-
2.1 Use of Combined Chemotherapy and Thoracic ably influence overall survival rates. The therapeutic
Radiation Therapy ..................................................... 136
ratio of radiation for the treatment of lung cancer can
2.2 Concurrent Therapy................................................... 136
2.3 Radiation Dose to the Thorax................................... 138 be improved by increasing the biological dose to
maintain local control while protecting normal
References.......................................................................... 139
tissues. One way of doing so is through the use of
fractionation, that is, manipulating the time interval
and dose of radiation to optimize the therapeutic ratio.
Topics covered in this chapter include dose escalation
with standard fractionation or hyperfractionation;
large-dose fractionation and stereotactic body radia-
tion therapy; accelerated fractionation; means of
reducing the target volume by using techniques such
as 3-dimensional conformal or intensity-modulated
radiation therapy; proton therapy; the importance of
accounting for tumor motion; and the use of combined
thoracic radiation and concurrent chemotherapy.

D. Gomez  M. D. Jeter  R. Komaki  J. D. Cox (&)


Division of Radiation Oncology, Unit 97,
The University of Texas MD Anderson Cancer Center, Radiation therapy has been an important component
1515 Holcombe Boulevard, Houston,
TX 77030, USA
of potentially curative treatment of lung cancer for
e-mail: jcox@mdanderson.org more than 40 years. The radiosensitivity of normal

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 129
DOI: 10.1007/174_2011_268, Ó Springer-Verlag Berlin Heidelberg 2011
130 D. Gomez et al.

Table 1 Fractionation definitions for lung cancer


Schedule Dose per No. fractions Intervals between Total no. Duration of Total dose
fraction (Gy) per week fractions (h) fractions treatment (weeks) (Gy)
Standard 1.8–2.75 4–6 24 25–40 5–8 55–75
Hypo [3.0 1–4 48–168a ; NC or ; NC or ;
Hyper 0.7–1.3 10–25 2–12 : (;) NC NC (:)
Rapid [2.5 5 24 ; ; ;
Accelerated 1.5–2.5 10–21 4–12 ; ;; ;
Numbers or symbols given assume a dose-rate of 2.0–6.0 Gy/min
; or : indicate decreases or increases relative to values given for standard fractionation schedule; NC indicates no change
a
Intervals longer than 168 h constitute a ‘‘split course.’’

tissues in the thorax, especially the normal lung and


esophagus, has led investigators to seek ways of 1 Non-Small Cell Lung Cancer
enhancing the antitumor effects of radiation while
reducing its acute and late adverse effects on normal Local control of NSCLC, like that of SCLC, is
tissues. The focus in this chapter is on medically directly related to survival. The ability to maintain
inoperable or locally advanced unresectable disease, local control of NSCLC has been far from satisfac-
specifically non-small cell lung cancer (NSCLC) tory, and hence several attempts have been made to
classified as stage IIIB (T4 or N3) or stage IIIA that is manipulate fractionation dose and schedule to esca-
unresectable because of bulky tumors or fixed N2 late the biologically effective dose to the tumor and
disease (according to the 2010 American Joint thus improve the outcome. Table 1 gives some defi-
Committee on Cancer staging system) and small cell nitions that are useful in reviewing the literature on
lung cancer (SCLC) confined to one hemithorax and time, dose, and fractionation in lung cancer.
the ipsilateral supraclavicular lymph nodes.
The success of radiation therapy in a particular set-
ting often depends on the endpoints used. The traditional 1.1 Dose Escalation with Standard
assumption has been that distant metastasis is the major Fractionation
cause of death from lung cancer; however, uncontrolled
tumor in the chest is also a major cause of mortality. The concept and standardization of dose constraints
Indeed, improvements in thoracic computed tomogra- for the lungs in the setting of standard fractionated
phy (CT) scanning and fiberoptic bronchoscopy that radiation therapy for lung cancer have changed in
allow better visualization of lung tumors have led to the recent decades. With regard to maximum tolerated
recognition that lack of local control is the main cause of doses, studies of fractionated irradiation delivered
treatment failure in lung cancer (Arriagada et al. 1991a, either to both lungs or to one lung at a time (Cox et al.
1997). Two independent randomized trials have shown 1972) suggested that an appropriate lung dose con-
that improving local control, which can be achieved straint was approximately 20 Gy at 1.5 Gy/fraction.
through two different approaches, can affect the overall With regard to the volume of lung subjected to radi-
survival rates in both SCLC and NSCLC (Saunders et al. ation, Graham et al. 1995 used dose–volume histo-
1997; Schaake-Koning et al. 1992). gram analysis to show that the percentage of normal
The therapeutic ratio of radiation for the treatment lung volume receiving a total dose of at least 20 Gy
of carcinoma of the lung can be improved by (V20) in standard daily fractions was strongly related
increasing the biological dose to maintain local con- to the risk of severe or life-threatening pulmonary
trol while protecting normal tissues. One way of toxicity.
doing so, and our emphasis in this chapter, is through The initial landmark study of dose escalation
the use of fractionation, i.e., manipulating the time with standard fractionation was conducted by the
interval and dose of irradiation to optimize the ther- Radiation Therapy Oncology Group (RTOG) (Perez
apeutic ratio. et al. 1988). Patients were randomly assigned to
Radiation Time, Dose, and Fractionation in the Treatment of Lung Cancer 131

one of four treatment groups. Three of these treat- 1.2 Large-Dose Fractionation
ments involved standard fractionation (2.0 Gy/day and Stereotactic Body Radiation
given 5 days/week) to total doses of 40 Gy (in 20 Therapy
fractions), 50 Gy (in 25 fractions), and 60 Gy
(in 30 fractions); the fourth treatment used large- Advocates of large-dose fractionation emphasize the
dose fractionation given in a split course (4 Gy/day usefulness of this form of treatment (also called
for 5 days, followed by a 3-week interruption, and a hypofractionation) in lessening the overall number of
second course of 4 Gy per day for 5 days) to a total treatments and the corresponding burden on health
dose of 40 Gy (in ten fractions over 5 weeks). The care facilities, reducing the stress on patients to
higher total radiation dose led to improved survival adhere to a schedule of 5 visits per week over a period
rates, but this effect came at the cost of increased of six weeks, and possibly in increasing the biological
toxicity. The RTOG investigators’ conclusion that antitumor effect. Thames et al. (1983) documented in
60 Gy in 30 fractions was the most effective the early 1980s that the use of large-dose fractions
treatment became the standard for the RTOG, for was associated with increased late effects in normal
other cooperative groups, and for radiation oncolo- tissues; a comprehensive review of large-dose frac-
gists throughout the US. tionation published 2 years later by Cox (1985) con-
However, since that time, other studies have firmed the increase in late effects but also drew
corroborated the feasibility of increasing the total attention to the possibility that this practice also had
dose while still using standard fractionation. In adverse effects on tumor control because it allowed
RTOG 9311, 177 patients receiving three- repopulation of tumor cells between fractions.
dimensional conformal radiation therapy (3D CRT) In a subsequent study involving large-dose frac-
were stratified to receive escalating radiation doses tionation, Uematsu et al. (2001) used CT-guided
at levels that depended on the V20. Twenty-five frameless stereotactic radiation therapy to treat 50
patients received induction chemotherapy, but no patients with stage I NSCLC. Most of the patients in
patient received concurrent chemotherapy. Patients this study were given 50–60 Gy in 5–10 fractions for
with a V20 of \25% received successive standard- 1–2 weeks, and 18 patients had already undergone
fractionation doses of 70.9, 77.4, 83.8 and 90.3 Gy. conventional radiation therapy (40–60 Gy in 20–33
Patients with V20 of 25–36% received successive fractions) before the stereotactic procedure. At a
doses of 70.9 and 77.4 Gy. The investigators found median follow-up time of 36 months, 47 patients
that radiation dose could be safely escalated with (94%) showed no evidence of local progression on
this technique to 83.8 Gy if the V20 was \25% and follow-up CT scans, and the 3-year overall survival
to 77.4 Gy if the V20 was between 25 and 36% rate was 66%. No adverse effects conclusively related
(Bradley et al. 2005). to the stereotactic radiation therapy were noted except
In the subsequent RTOG 0117 trial, radiation dose for minor bone fractures (two patients) and temporary
escalation with 3D CRT was studied in a phase I/II pleural pain (six patients).
setting with concurrent paclitaxel and carboplatin During the past decade, the use of stereotactic
chemotherapy. In the initial study design, the radia- body radiation therapy (SBRT) has increased sharply.
tion dose was to be intensified by increasing the daily Improved imaging techniques such as cone-beam CT
fraction size, starting from a total dose of 75.25 Gy to have allowed precise localization of the tumor during
be delivered in 35 fractions. However, among the treatment that minimizes treatment margins and sub-
initial eight patients, two experienced dose-limiting sequent toxicity from normal tissue damage. One of
toxicity and thus, the protocol was revised to the initial reports on the safety and efficacy of this
de-escalate the radiation dose. The investigators technique for early-stage lung tumors was from the
reported that the maximum tolerated dose was 74 Gy University of Indiana. In that investigation, 37
in 37 fractions (Bradley et al. 2010). These results patients with T1 or T2 disease received SBRT at an
led to an RTOG randomized trial that is currently initial dose of 8 Gy/fraction, with successive dose
ongoing (RTOG 0617) comparing total doses of 60 to escalations of 2 Gy/fraction. A dose of 60 Gy was
74 Gy with concurrent chemotherapy for local- achieved for both patients with T1 and those with T2
regionally advanced NSCLC. disease, and no failures were noted among patients
132 D. Gomez et al.

who had received at least 18 Gy per fraction fractions were given twice daily and the total doses
(Timmerman et al. 2003). A subsequent study from were escalated (Diener-West et al. 1991). For cancer of
the same institution demonstrated that the rate of the lung, the total doses ranged from 60 Gy (in 50
severe toxicity for centrally located tumors, defined as fractions over 5 weeks) to 79.2 Gy (in 66 fractions over
those within 2 cm of the bronchial tree, approached 6.5 weeks). Improved survival rates were noted at the
50% (Timmerman et al. 2006), prompting recom- total dose of 69.6 Gy, given in 58 fractions, with no
mendations that lower fractionation regimens be used further improvement at higher doses. This dose frac-
for tumors in this location. tionation regimen was subsequently investigated in a
A recent report of 59 patients with solely periph- prospective trial in comparison with groups given either
eral tumors treated with 54 Gy in 3 fractions (18 Gy standard fractionation or induction chemotherapy
per fraction) demonstrated a primary tumor control followed by standard fractionation (Sause et al. 1995).
rate of 97.6% and a local-regional control rate of In that trial, use of induction chemotherapy was
87.2% at a median follow-up time of 34.4 months. associated with improved short-term survival but use
Twelve percent of patients experienced grade 3 tox- of the hyperfractionated regimen was not.
icity, and two patients experienced grade 4 toxicity,
but none experienced grade 5 toxicity (Timmerman
et al. 2010). Although this local control rate is 1.4 Accelerated Fractionation
promising, longer follow-up is needed to establish the
effectiveness of SBRT in the long term. In the With its twice-daily doses of 1.2 Gy, the protocol in
meantime, the RTOG is conducting two trials, both of the RTOG study cited in the previous paragraph did
which were ongoing when this chapter was written. involve some acceleration of treatment; however, the
RTOG 0915 is a randomized phase II study compar- most thorough investigation of a markedly acceler-
ing SBRT schedules for patients with stage I periph- ated course of radiation therapy was conducted at
eral NSCLC who are not candidates for surgery. Mount Vernon Hospital in the UK by Saunders et al.
Patients are randomly assigned to one of two frac- (Saunders et al. 1997; Saunders 2000). Their inves-
tionation regimens, either 48 Gy in four once-daily tigation of continuous hyperfractionated accelerated
fractions or 34 Gy in one fraction. The primary end- radiation therapy (CHART) involved use of three
point is the rate of grade 3 or higher toxicity at 1 year. 1.5 Gy fractions per day for a total of 12 consecutive
The other study, RTOG 0813, was designed to treatment days, with no interruptions for weekends.
identify the maximum tolerated dose for patients with The total dose for this regimen was 54 Gy, given in
centrally located, T1/T2N0M0 NSCLC who are not 36 fractions over 12 days.
candidates for surgical resection. Doses are escalated After CHART was found to produce promising
beginning at 50 Gy in five fractions, with the target results in comparison with the historical experience at
dose being 60 Gy in five fractions. A secondary Mount Vernon Hospital, a prospective randomized
endpoint is the local control rate at the maximum trial was undertaken to compare CHART (total dose
tolerated dose. of 54 Gy given in 36 fractions over 12 consecutive
days) to standard fractionation (total dose of 60 Gy in
30 fractions, 5 days per week, for 6 weeks). The
1.3 Dose Escalation observed improvement in survival in the CHART
with Hyperfractionation group was considered to result largely from the
improved intrathoracic tumor control. A derivative
The potential for hyperfractionation to improve the benefit of this local tumor control was the lesser
therapeutic ratio of radiation for many malignant incidence of distant metastasis in the CHART group
tumors was recognized in part from the failure of compared with the standard-fractionation group
large-dose fractionation to improve local control and (Saunders, 2000). This finding suggests that metas-
in part from the observation that use of smaller frac- tasis from locally advanced lung cancer, like that at
tions was associated with fewer late effects in normal other cancer sites, may arise through secondary
tissues. The RTOG conducted a series of trials of dissemination from residual local-regional tumors
hyperfractionated radiation therapy in which 1.2-Gy (Arriagada et al. 1995).
Radiation Time, Dose, and Fractionation in the Treatment of Lung Cancer 133

Several reports have appeared during the past with disease rather than broadly applying nodal irra-
5 years on the use of hyperfractionation for NSCLC. diation. Several investigators have shown that
Jeremic and Milicic (2008) examined the results of 78 involved-field irradiation is feasible with failure rates
patients with stage I or stage II NSCLC treated with between 5 and 10% (Rosenzweig et al. 2001, 2007).
either conventionally fractionated or hyperfrac- The involved-field technique allows doses to be
tionated radiation, and they found that a hyperfrac- escalated to regions at risk while minimizing the dose
tionated regimen resulted in slightly improved overall to normal structures; by avoiding broadly applied
survival with similar toxicity (Jeremic and Milicic elective nodal irradiation for locally advanced lung
2008). Din et al. (2008) examined 583 patients with cancer, some investigators have been able to increase
stage I–IV disease who had received CHART from the radiation dose to the tumor above 80 Gy
1997 to 2005 at five centers in the UK and found that (Belderbos et al. 2003). A group at the Shandong
only four cases of grade 4 or 5 toxicity had been Cancer Hospital in China conducted a randomized
documented. The median survival time, 16.2 months, trial to compare elective nodal irradiation to involved-
was comparable to that in the original study exam- field irradiation as part of definitive chemoradiation
ining this technique. The current practice at MD for inoperable stage III NSCLC. These investigators
Anderson Cancer Center is to reserve the use of concluded that use of involved-field irradiation could
hyperfractionated regimens for tumors located near increase the overall survival time, improve local
neurologic structures such as the spinal cord and control, and reduce rates of radiation pneumonitis
brachial plexus, because sparing these structures from (Yuan et al. 2007). However, this trial must be
late effects after high doses of standard fractionated interpreted cautiously because of its single-institution
radiation is difficult to accomplish. Such tumors are design and its use of different doses in the two
generally treated with twice-daily 1.2 Gy fractions to treatment arms, with the involved-field irradiation
a total dose of 69.6 Gy. arm receiving a higher tumor dose. A subsequent
editorial on elective nodal irradiation that included
investigators from several institutions highlighted the
1.5 Reducing the Target Volume with 3D fact that achieving effective involved-field irradiation
Conformal Radiation Therapy depends largely on the confidence of the treating
physician in the quality of the diagnostic imaging.
Some evidence exists to suggest that very high radia- Without the benefit of positron emission tomography
tion doses (i.e., in excess of 70 Gy) for medically (PET) scanning or appropriate mediastinal staging
inoperable stage I disease can produce acceptable local methods (bronchoscopy, mediastinoscopy), defining
control and survival (Qiao et al. 2003). Patients with ‘‘involved’’ nodes can be difficult if not impossible.
small but inoperable tumors may be candidates for 3D The authors of the editorial concluded that clinicians
CRT, which can allow dose escalation if the high-dose should adopt a ‘‘tailored’’ approach that would reflect
radiation volume conforms closely to the size and shape the range of imaging and investigational tools that
of the tumor. Essential components of 3D CRT include are available for assessing an individual patient’s
the capability for CT simulation, beam’s-eye-view case such that selective nodal irradiation could be a
treatment planning, and multileaf collimators to possibility in some cases (Belderbos et al. 2009).
maximize conformality around the treatment volume.
The relationship between pulmonary toxicity, espe-
cially symptomatic toxicity, and the volume of irradi- 1.6 Intensity-Modulated Radiation
ated lung is well established, and dose escalation has Therapy
been performed successfully in this setting to doses
approaching 85 Gy. Indeed, conformal techniques Development of sophisticated software programs,
have become the standard of care in the treatment of combined with improvements in diagnostic imaging
lung cancer because they can spare structures such as and image reconstruction, have allowed tumors to be
the lung, esophagus, heart, and spinal cord. visualized and delineated ever more precisely, which
By using 3D CRT techniques, it has been possible improves the delivery of 3D CRT and opens the door
to selectively target lymph nodes that are involved for intensity-modulated radiation therapy (IMRT).
134 D. Gomez et al.

Two sources of error in IMRT that could prevent the tumor motion and location may be required to ensure
successful delivery of optimal conformal treatment that tumors remain within the high-dose region
result from movement—movement of the patient, throughout the course of treatment.
which can be addressed by careful immobilization, At MD Anderson Cancer Center, treatments for all
and the internal motion of thoracic tumors caused by patients are simulated while the patients hold their arms
respiration and heartbeats. Indeed, the main challenge over their head to maximize the number of potential
in IMRT is to avoid increasing the integral dose to beam arrangements. All patients undergo 4D CT during
organs that lie in the path of the multiple fields that which an external fiducial is placed on the abdomen to
are focused on the target volume. assess tumor motion in terms of respiratory excursion.
In practice, the question to be considered in the If the tumor is found to move up to 1 cm in any direc-
choice of treatment technique for thoracic tumors is tion, the patient is treated with a ‘‘free-breathing’’
whether IMRT can reduce the V20. Several dosimetric technique, in which the extent of tumor motion is
reports have shown that IMRT can indeed reduce lung accounted for by using a single expanded target
dose compared with 3D CRT for locally advanced volume, the internal target volume. However, if the
NSCLC (Christian et al. 2007; Grills et al. 2003). tumor moves more than 1 cm and the patient is capable
A 2010 report from MD Anderson Cancer Center of controlling their breathing, then radiation is deliv-
compared the outcomes of 496 patients who had been ered while the patient holds their breath at full inspi-
treated between 1999 and 2006 with concomitant ration (deep inspiratory breath hold technique), or the
chemotherapy and either CT-based treatment radiation is timed for delivery at a chosen point in
planning with 3D CRT or 4D CT-based planning and the breathing cycle (ventilator gated technique).
IMRT. The comparison showed that both the V20 and
the rate of severe pneumonitis were significantly
lower in the IMRT group. The overall survival was 1.8 Proton Therapy
also improved in the patients who received 4D CT
with IMRT. Rates of disease-free survival and distant Proton therapy represents a new paradigm in the cate-
metastases were similar among the two groups (Liao gory of conformal therapy. The advantage of proton
et al. 2010). These results suggest that if the resources beam therapy lies in the dose distribution characteris-
are available, IMRT should be adopted as the stan- tics of the proton particles; protons deliver nearly all
dard practice for locally advanced NSCLC. their energy at the height of the Bragg peak, meaning
that any exit dose past the target is negligible. During
the past 5 years, results from several dosimetric com-
1.7 Accounting for Tumor Motion parisons of proton therapy with other techniques have
been published, and early clinical outcomes after
That intrathoracic tumors move in concert with respi- proton therapy for lung cancer are becoming available.
ratory and cardiac cycles has always been known, but Chang et al. (2006) published dosimetric results of
the importance of accounting for this movement has patients with stage I or III NSCLC treated with 3D
been magnified with the advent of 3D CRT and IMRT. CRT, IMRT, or proton therapy. The authors found that
Several studies have been performed during the past all three techniques could provide excellent target
decade to characterize the direction and extent of lung coverage, but doses to the lung, spinal cord, esophagus,
tumor movement. Although tumors can move signifi- and integral dose were all lower with proton therapy
cantly in any direction, the largest movements most than with the other two techniques (Chang et al. 2006).
often occur in the superior–inferior direction (Michalski These dosimetric advantages seem to be translating
et al. 2008), and small tumors tend to move more than improved clinical outcomes. Sejpal et al. (2011) com-
larger tumors (Liu et al. 2007). Moreover, because pared the toxicity of three radiation therapy techniques,
the location and size of the tumor can change over the all in combination with concurrent chemotherapy, for
course of radiation therapy that lasts several weeks, the NSCLC. At a median follow-up time of approximately
importance of recognizing and accounting for tumor 16 months, the median radiation dose from proton
motion over the course of radiation therapy is becoming therapy was found to be higher than the doses delivered
increasingly apparent. Hence repeated monitoring of by IMRT or 3D CRT (74 Gy (RBE) vs. 63 Gy,
Radiation Time, Dose, and Fractionation in the Treatment of Lung Cancer 135

respectively), but the proton therapy produced lower Organisation for Research and Treatment of Cancer
rates of severe pneumonitis and esophagitis than did (Schaake-Koning et al. 1992) that showed improved
IMRT or 3D CRT (Sejpal et al. 2011). Chang et al. local control and overall survival from the use of
(2011) published early results of a prospective phase I/II concurrent chemotherapy and radiation. However,
trial examining the efficacy of high-dose, slightly this trial was based on a fractionation scheme that
hypofractionated proton therapy for medically inopera- may have been less than optimal, in that 3-Gy frac-
ble T1N0 or T2-T3N0 NSCLC. All patients received a tions were given daily for 10 days, followed by an
dose of 87.5 Gy in 2-Gy fractions. At a median follow- interruption of 4 weeks and then daily 2.5-Gy frac-
up time of 16.3 months, the rate of local control was tions for 10 days. Randomized studies of fractionated
88.9%, and no patient had experienced any grade 4 or radiation for locally advanced carcinomas of the
grade 5 toxicity. Notably, no patient had experienced upper respiratory and digestive tract (Fu et al. 2000)
grade 3 or higher pneumonitis (Chang et al. 2011). A suggest that interruptions such as this are quite likely
meta-analysis comparing the effectiveness of photons, to have allowed the proliferation of surviving clono-
protons, and carbon-ion therapy for NSCLC showed that gens, not only in normal tissues but also in the tumor.
survival rates for the patients treated with protons or Also, a recent meta-analysis based on individual
carbon ions were higher than those for patients who patient data raised some doubts about the magnitude
underwent photon therapy when standard fractionation of benefit (Auperin and Le Pechoux 2003) from
schemes were used. Survival rates were similar to those concurrent chemotherapy and radiation therapy and
obtained from SBRT with photons (Grutters et al. 2010). suggested that additional randomized evidence was
Virtually all studies that have been published on needed to support use of the combined approach.
proton therapy for NSCLC have involved the use of a The most extensive experience with using altered
passive scattering technique. However, some evi- fractionation with concurrent chemotherapy for
dence exists to suggest the feasibility of modulating NSCLC comes from MD Anderson Cancer Center
proton fluence, akin to the modulation of photons in and the RTOG. Results from randomized phase II
IMRT. A dose–volume histogram analysis of treat- trials of a fractionation scheme developed by the
ment plans generated for intensity-modulated proton RTOG (58 twice-daily 1.2-Gy fractions for a total
therapy, for IMRT, or for passive scattering proton dose of 69.6 Gy) used with concurrent chemotherapy
therapy of locally advanced NSCLC indicated that seemed promising (Komaki et al. 1997). However,
intensity-modulated proton therapy spared the lung, the most favorable outcomes seemed to be associated
heart, spinal cord, and esophagus to a great extent with a learning curve; specifically, institutions at
than did IMRT. Intensity-modulated proton therapy which five or more patients received concurrent
further allowed an increase in the maximum tolerated chemotherapy and twice-daily irradiation showed
dose from 74 Gy with passive scattering proton significantly better survival rates than institutions
therapy to 84.4 Gy without significant compromise in with less experience in this form of treatment (Lee
normal tissue doses (Zhang et al. 2010). If intensity et al. 2002). At MD Anderson Cancer Center, long-
modulation of proton therapy could be successfully term follow-up of patients given 1.2-Gy fractions
achieved in clinical practice, this planning technique twice a day with concurrent cisplatin and etoposide
would offer an additional advantage over IMRT by showed a 5-year survival rate of 26%—the most
allowing range modulation as well, which cannot be favorable such rate reported to date (Liao et al. 2002).
done with photon techniques. Future studies will offer
further information on the clinical applicability of
intensity-modulated proton therapy. 2 Small Cell Lung Cancer

The current treatment strategy for limited-stage SCLC


1.9 Concurrent Chemotherapy involves the use of chemotherapy, thoracic radiation
therapy (Turrisi et al. 1999), and, for those who
The use of concurrent weekly or daily cisplatin with achieve a complete response, prophylactic cranial
radiation therapy has been based mainly on the irradiation (PCI) (Auperin et al. 1999). Comparisons
results of a large randomized trial by the European of chemotherapy plus thoracic radiation therapy with
136 D. Gomez et al.

chemotherapy alone have shown that use of combi- inability to assess response to either modality; and
nation therapy improves survival rates; other trials possibly sensitization of normal tissues.
have shown that concurrent chemotherapy and tho- In 1990, McCracken et al. (1990) reported the
racic radiation therapy is superior to sequential or results of a phase II trial of the Southwest Oncology
alternating chemotherapy and thoracic radiation Group in which two courses of cisplatin, etoposide,
therapy with regard to local-regional control and and vincristine were given concurrently with radiation
survival for patients with limited-stage SCLC. therapy consisting of once-daily 1.8 Gy fractions
given 5 days/week to a total dose of 45 Gy. The
concurrent therapy was followed by additional
2.1 Use of Combined Chemotherapy chemotherapy with vincristine, methotrexate, and
and Thoracic Radiation Therapy etoposide alternating with doxorubicin and cyclo-
phosphamide for 12 weeks. This study evaluated 154
Because even initially localized SCLC tends to patients. With a minimum observation period of
metastasize early in the course of the disease, 3 years, the 2-year survival rate was 42% and the
chemotherapy is an essential component of the 4-year survival rate was 30%. An updated analysis
treatment regimen; intrathoracic failure becomes (Janaki et al. 1994) after a longer observation period
more important after distant metastases are con- showed a 5-year survival rate of 26%.
trolled. Two separate meta-analyses have confirmed In 1999, the RTOG and the Eastern Cooperative
the value of adding thoracic radiation therapy to Oncology Group (ECOG) (Turrisi et al. 1999)
chemotherapy for SCLC in terms of decreasing the reported the results of a US nationwide randomized
rate of local recurrence and improving survival. study of limited-stage SCLC treated with concurrent
Warde and Payne (1992) analyzed results from 11 chemotherapy (etoposide and cisplatin) and thoracic
prospective randomized trials of chemotherapy with radiation therapy (45 Gy given in twice-daily 1.5-Gy
or without thoracic radiation therapy for patients with fractions or once-daily 1.8 Gy fractions); radiation
limited-stage SCLC and found that the addition of was begun on the first day of the chemotherapy cycle.
thoracic radiation therapy conferred an absolute The 2-year survival rate for the entire group was 44%.
increase of 5.4% in the overall survival rate at 2 years The 5-year survival rate was 16% for those given
(from 15 to 20.4%) and an absolute increase of 25% once-daily radiation and 26% for those given twice-
in the local control rate at 2 years (from 15 to 40%). daily radiation—a remarkable improvement over
Pignon et al. (1992), in their analysis of data from previously reported 5-year survival rates.
2,140 patients in 13 randomized trials of chemother- The Japanese Clinical Oncology Group (Goto et al.
apy alone versus chemotherapy plus thoracic radia- 1999) conducted a phase III study of concurrent
tion therapy, found an absolute increase of 5.4% in versus sequential thoracic radiotherapy, given in
the overall survival rate at 3 years. combination with cisplatin and etoposide chemo-
therapy, for patients with limited-stage SCLC.
Chemotherapy was given in either a 28-day cycle (the
2.2 Concurrent Therapy concurrent group) or a 21-day cycle (the sequential
group). Thoracic radiation therapy was begun either
Potential advantages of delivering chemotherapy and on day 2 of the first cycle of chemotherapy in the
radiation therapy concurrently are the ability to apply concurrent group or after the fourth cycle of chemo-
both modalities early in the course of treatment; the therapy in the sequential group. The radiation therapy
possible induction of synergistic effects; the enhanced consisted of 45 Gy delivered to the thorax in twice-
accuracy of treatment planning (because induction daily 1.5-Gy fractions over 3 weeks. PCI was given to
chemotherapy may obscure the original tumor volume); patients who showed a complete or a near-complete
and the short overall treatment time (high dose- response; the PCI consisted of 24 Gy given in twice-
intensity), which prevents the proliferation of clono- daily 1.5-Gy fractions given 5 days a week. The inci-
gens. Potential disadvantages of concurrent therapy are dence of grade 3 or 4 leukopenia was significantly
enhanced toxicity to normal tissues, which could higher in the concurrent-therapy group (86.8 vs. 51.3%,
necessitate dose modification or treatment breaks; the P \ 0.001), but the incidence of non-hematologic side
Radiation Time, Dose, and Fractionation in the Treatment of Lung Cancer 137

effects was no different in the two groups. The 2- and study, patients with limited-stage SCLC received six
3-year survival rates in the sequential-therapy group cycles of the same alternating CAV/PE chemotherapy
were 35.4 and 20.7%, respectively, as compared and were then assigned to receive radiation either
with 55.3 and 30.9% in the concurrent-therapy group. early (at the second chemotherapy cycle) or late
Overall survival seemed to be superior in the (at the sixth chemotherapy cycle). The radiation reg-
concurrent group, but this apparent trend was not imen was also the same as that in the Canadian trial,
statistically significant. 40 Gy in 15 fractions, and patients with a response
Arriagada et al. (1991a, b) reported the results of were offered PCI, also 25 Gy in ten fractions. The
two protocols involving 72 consecutive patients with authors found no difference in survival between the
limited-stage SCLC. Patients were given two cycles groups given radiation early or late in the course of
of induction chemotherapy followed by three 2-week chemotherapy (Spiro et al. 2006).
cycles of thoracic radiation therapy that included To reconcile the results of randomized trials such
chemotherapy with the same regimen as that used for as these, a meta-analysis was published in 2007
the induction. Cisplatin and etoposide were used in comparing early versus late radiation therapy.
the first trial, and cisplatin, etoposide, cyclophospha- ‘‘Early’’ was defined as within 30 days after the start
mide, and doxorubicin were used in the second trial. of chemotherapy, and the results were analyzed
The results of this trial are among the most favorable according to the treatment time and the use of plati-
reported in terms of long-term survival. The complete num-based chemotherapy regimens. When all seven
response rate was 87% and the overall survival rate randomized trials were examined, no clear benefit was
was 26% at 3 years; the overall survival of patients found regarding early versus late radiation. However,
who showed a complete response to the interdigitated when only those trials were included in which patients
therapy was 26% at 5 years. received chest radiation over a period lasting less than
Whether thoracic radiation therapy should be 30 days, a statistically significant improvement was
delivered early or late in the treatment course remains noted in the overall survival. Similarly, when only
controversial. The National Cancer Institute of Canada those studies involving platinum chemotherapy regi-
Clinical Trials Group studied this issue in a randomized mens were included, a slight benefit in the 5-year
trial (Murray et al. 1993). In that trial, 308 patients were overall survival rate was found with early RT
given six cycles of chemotherapy with cyclophospha- (Pijls-Johannesma et al. 2007). Based on results such
mide, doxorubicin, and vincristine alternating with as these, the standard practice at MD Anderson
etoposide and cisplatin (CAV/PE). Patients were ran- Cancer Center is to begin thoracic radiation during the
domly assigned to receive thoracic radiation therapy first or second cycle of chemotherapy. This practice
(40 Gy to the primary tumor site in 15 fractions over is also supported by the National Comprehensive
3 weeks given concurrently with etoposide and cis- Cancer Network.
platin) beginning either at week 3 (the early group) or at
week 15 (the late group). Those who showed a com- 2.2.1 Fractionation
plete response were then given PCI (25 Gy in ten Three fractionation regimens have been shown to be
fractions over 2 weeks) after the completion of all effective for limited-stage SCLC. First, in the widely
chemotherapy and thoracic irradiation. Although the cited Intergroup study 0096 (Turrisi et al. 1999)
complete response rates were no different in the two conducted with the ECOG and RTOG, investigators
groups, progression-free survival (P = 0.036) and compared once-daily versus twice-daily radiation
overall survival (P = 0.008) were significantly better therapy in combination with concurrent cisplatin and
in the early-radiation group. Patients in the late- etoposide. All patients received four 21-day cycles of
radiation group also had a significantly higher rate of chemotherapy. The once-daily fractionation group
brain metastasis (P = 0.006). This study indicated that received a single 1.8-Gy fraction each day, to a total
early use of thoracic radiation therapy with concurrent dose of 45 Gy in 25 fractions over 5 weeks. The
chemotherapy improved survival, possibly by elimi- twice-daily fractionation group received two 1.5-Gy
nating the clonogens in the primary tumor. fractions each day, with a 4- to 6-h interval between
However, this study was essentially repeated by fractions, to a total dose of 45 Gy in 30 fractions
investigators in the UK with conflicting results. In that over 3 weeks. Irradiation began during the first
138 D. Gomez et al.

chemotherapy cycle. Patients who achieved a com-


plete response then were offered PCI (ten 2.5-Gy
fractions). Although accelerating the radiation
improved median survival time (from 19 months for
the standard fractionation group to 23 months for the
twice-daily group) and 2-year survival rates (41 vs.
47%), a statistically significant difference in survival
was not apparent until 5 years (16 vs. 26%;
P = 0.04). The accelerated regimen also produced
acute grade 3 esophagitis in 27% of cases as
compared with 11% of those in the once-daily
fractionation group.
The second regimen was tested by the Cancer and
Leukemia Group B (CALGB), which studied 63
Fig. 1 Treatment Arms in CALGB 30610/RTOG 0538:
patients with limited-SCLC who received induc- BID = twice daily, fx = fraction
tion chemotherapy with paclitaxel and topotecan,
followed by three cycles of carboplatin and etoposide
concurrent with thoracic radiation to 70 Gy in 2-Gy the total doses given were 45 Gy (i.e., doses split
fractions. PCI was offered to patients experiencing a 15–15–15), 55 Gy (20–20–15), and 65 Gy (20–20–
complete or partial response. The 2-year overall sur- 25). The corresponding 3-year local control rates were
vival rate in this study was 48%, and the progression- 66% for the group given 45 Gy and 70% for the two
free survival rate was 31%. Thus the rates of overall higherdose groups; the 5-year survival rates were 16%
survival and disease-free survival as well as those of for the 45-Gy group, 16% for the 55-Gy group, and
hematologic and esophageal toxicity were similar in 20% for the 65-Gy group. None of these apparent
the CALGB and INT 0096 trials (Bogart et al. 2004). differences were statistically significant among the
The third regimen, evaluated in RTOG 0239, three groups. The overall incidence of lethal toxicity
consisted of 61.2 Gy delivered in 5 weeks delivered was 10%, and this rate was no different among any of
with the first two of four cycles of etoposide and the three radiation dose groups.
cisplatin. The first 16 fractions were given once daily, Choi et al. (1998) conducted a phase I study to
and after 16 fractions a field reduction was made determine the maximum tolerated dose of radiation
based on an adaptive treatment simulation, at which given either in standard daily fractionation or
time the regimen was changed to twice daily. The in hyperfractionated-accelerated twice-daily radiation
findings from that trial have yet to be published, schedules with concurrent chemotherapy for limited-
although this regimen is currently being compared stage SCLC. The maximum tolerated dose of hyper-
with the first two in a joint study (CALGB 30610/ fractionated radiation therapy was 45 Gy given in
RTOG 0538) (Fig. 1). PCI is offered in all patients 30 fractions over 19 days. However, in daily fraction-
with a complete or near-complete response. The pri- ation, the maximum tolerated dose was not reached at
mary endpoint is overall survival, and secondary 66 Gy given in 33 fractions over 45 days, and thus
endpoints are complete response rates, progression- patients were accrued for a third group to receive 70 Gy
free survival, and tumor progression. in 35 fractions over 47 days. The tumor response rates
varied from 78 to 100%, and no difference was found
among dose levels. Doses above 40 Gy did not
2.3 Radiation Dose to the Thorax significantly improve the local control rate. Esophagitis
and granulocytopenia of grade 3 or higher were more
Arriagada et al. (1990) at the Institut Gustave-Roussy common among patients given the hyperfractionated
conducted three consecutive trials of 173 patients and accelerated-fractionation treatments.
with limited-stage SCLC treated with different To clarify the maximum tolerated dose of thoracic
thoracic radiation doses. All thoracic radiation was radiation (in terms of acute esophagitis and pneu-
given in split courses alternating with chemotherapy; monitis) that could be given in combination with
Radiation Time, Dose, and Fractionation in the Treatment of Lung Cancer 139

Table 2 Intergroup Study 0096 versus RTOG 9712


Intergroup RTOG 9712

Group 1 Group 2
Thoracic radiation dose 45 Gy 45 Gy 61.2 Gy
Duration of radiation 5 weeks 3 weeks 5 weeks
Median survival time 19 months 23 months –
Survival rates
1-year 63% 67% –
2-years 44% 47% –
5-years 16% 26%* –
Local failure rate 52% 36% –
Incidence of grade 3 esophagitis 11% 27% \40%
*Significantly different from Group 1 (P = 0.01)

cisplatin and etoposide chemotherapy for patients with chemotherapy on locally advanced non-small cell lung
limited-stage SCLC, the RTOG conducted trial 9712 carcinoma: a randomized study of 353 patients. Int J Radiat
Oncol Biol Phys 20(6): 1183–1190
(Table 2) (Komaki et al. 2003). The findings of this Arriagada R, Le Chevalier T, Quoix E, Ruffie P, de Cremoux H,
phase I trial indicated that doses could be escalated to Douillard JY et al (1991b) ASTRO (American Society for
61.2 Gy over 5 weeks through the use of a concomi- Therapeutic Radiology and Oncology) plenary: effect of
tant boost technique without more than 40% of patients chemotherapy on locally advanced non-small cell lung
carcinoma: a randomized study of 353 patients. GETCB
developing esophagitis of grade 3 or higher. This total (Groupe d’Etude et Traitement des Cancers Bronchiques),
dose was given as follows. Eleven 1.8-Gy fractions FNCLCC (Federation Nationale des Centres de Lutte contre
were given to large fields once daily for 5 days a week, le Cancer) and the CEBI trialists. Int J Radiat Oncol Biol
followed by 4 days of twice-daily radiation therapy in Phys, 20(6): 1183–1190
Arriagada R, Rutqvist LE, Mattsson A, Kramer A, Rotstein S
which one 1.8-Gy fraction was given in the morning to (1995) Adequate locoregional treatment for early breast
large fields and another 1.8-Gy fraction was delivered cancer may prevent secondary dissemination. J Clin Oncol
to boost fields 6 h later; for the final 5 days, twice- 13(12):2869–2878
daily 1.8-Gy fractions were given to the boost fields. Arriagada R, LeChevalier T, Rekacewicz C, Quoix E, De
Cremoux H, Douillard JY et al (1997) Cisplatin-based
This regimen is currently being compared to two chemotherapy (CT) in patients with locally advanced non-
alternative regimens in the protocol CALGB 30610/ small cell lung cancer (NSCLC): late analysis of a French
RTOG 0538, as described above. randomized trial. Proc ASCO 16:446a
Auperin A, Le Pechoux C (2003) Meta-analysis of randomized
Acknowledgment Supported in part by National Cancer trials evaluating cisplatin or carboplatin-based concom-
Institute grants CA 16672 and 06294 and the Texas Tobacco itant chemoradiation versus radiotherapy alone in locally
Settlement. advanced non-small cell lung cancer. Paper presented at the
10th World Conference on Lung Cancer, Vancouver,
Canada
Auperin A, Arriagada R, Pignon JP, Le Pechoux C, Gregor A,
References Stephens RJ et al (1999) Prophylactic cranial irradiation for
patients with small-cell lung cancer in complete remission.
Prophylactic Cranial Irradiation Overview Collaborative
Arriagada R, le Chevalier T, Ruffie P, Baldeyrou P, De Group. N Engl J Med 341(7):476–484
Cremoux H, Martin M et al (1990) Alternating radiotherapy Belderbos JS, De Jaeger K, Heemsbergen WD, Seppenwoolde
and chemotherapy in 173 consecutive patients with limited Y, Baas P, Boersma LJ et al (2003) First results of a phase I/
small cell lung carcinoma. GROP and the French Cancer II dose escalation trial in non-small cell lung cancer using
Center’s Lung Group. Int J Radiat Oncol Biol Phys 19(5): three-dimensional conformal radiotherapy. Radiother Oncol
1135–1138 66:119–126
Arriagada R, Le Chevalier T, Quiox E, Ruffie P, De Cremoux Belderbos JS, Kepka L, Kong FM, Martel MK, Videtic GM,
H, Douillard JY et al (1991a) ASTRO plenary: effect of Jeremic B (2009) Elective nodal irradiation (ENI) in locally
140 D. Gomez et al.

advanced non-small-cell lung cancer (NSCLC): evidence carcinomas: first report of RTOG 9003. Int J Radiat Oncol
versus opinion? Int J Radiat Oncol Biol Phys 74(1):322 Biol Phys 48(1):7–16
author reply 322–323 Goto K, Nishiwaki Y, Takada M et al (1999) Final results of a
Bogart JA, Herndon JE II, Lyss AP, Watson D, Miller AA, Lee phase III study of concurrent versus sequential thoracic
ME et al (2004) 70 Gy thoracic radiotherapy is feasible radiotherapy (TRT) in combination with cisplatin (P) and
concurrent with chemotherapy for limited-stage small-cell etoposide (E) for limited-stage small cell lung cancer (LD-
lung cancer: analysis of Cancer and Leukemia Group B SCLC): the Japan Clinical Oncology Group (JCOG) study
study 39808. Int J Radiat Oncol Biol Phys 59(2):460–468 (abstract). Proc ASCO 18:A1805
Bradley J, Graham MV, Winter K, Purdy JA, Komaki R, Roa Graham MV, Purdy JA, Emami B, Matthews JW, Harms WB
WH et al (2005) Toxicity and outcome results of RTOG (1995) Preliminary results of a prospective trial using three
9311: a phase I–II dose-escalation study using three- dimensional radiotherapy for lung cancer. Int J Radiat
dimensional conformal radiotherapy in patients with Oncol Biol Phys 33:993–1000
inoperable non-small-cell lung carcinoma. Int J Radiat Grills IS, Yan D, Martinez AA, Vicini FA, Wong JW, Kestin LL
Oncol Biol Phys 61(2):318–328 (2003) Potential for reduced toxicity and dose escalation in the
Bradley JD, Moughan J, Graham MV, Byhardt R, Govindan R, treatment of inoperable non-small-cell lung cancer: a com-
Fowler J et al (2010) A phase I/II radiation dose escalation parison of intensity-modulated radiation therapy (IMRT), 3D
study with concurrent chemotherapy for patients with conformal radiation, and elective nodal irradiation. Int J
inoperable stages I to III non-small-cell lung cancer: phase Radiat Oncol Biol Phys 57(3):875–890
I results of RTOG 0117. Int J Radiat Oncol Biol Phys Grutters JP, Joore MA, Wiegman EM, Langendijk JA, de
77(2):367–372 Ruysscher D, Hochstenbag M et al (2010) Health-related
Chang JY, Zhang X, Wang X, Kang Y, Riley B, Bilton S et al quality of life in patients surviving non-small cell lung
(2006) Significant reduction of normal tissue dose by proton cancer. Thorax 65(10):903–907
radiotherapy compared with three-dimensional conformal or Janaki L, Rector D, Turrisi A et al (1994) Patterns of failure and
intensity-modulated radiation therapy in Stage I or Stage III second malignancies from SWOG-8629: concurrent cis-
non-small-cell lung cancer. Int J Radiat Oncol Biol Phys platin, etoposide, vincristine, and once daily radiotherapy
65(4):1087–1096 for the treatment of limited small cell lung cancer (abstract).
Chang JY, Komaki R, Wen HY, De Gracia B, Bluett JB, Proc ASCO 13:331
McAleer MF et al (2011) Toxicity and patterns of failure of Jeremic B, Milicic B (2008) From conventionally fractionated
adaptive/ablative proton therapy for early-stage, medically radiation therapy to hyperfractionated radiation therapy alone
inoperable non-small cell lung cancer. Int J Radiat Oncol and with concurrent chemotherapy in patients with early-
Biol Phys stage nonsmall cell lung cancer. Cancer 112(4):876–884
Choi NC, Herndon JE II, Rosenman J, Carey RW, Chung CT, Komaki R, Swann RS, Ettinger D (2003) Phase I study of
Bernard S et al (1998) Phase I study to determine the thoracic radiation dose-escalation with concurrent chemo-
maximum-tolerated dose of radiation in standard daily and therapy for patients with limited small cell lung cancer
hyperfractionated-accelerated twice-daily radiation sched- (LSCLC): Radiation Therapy Oncology Group (RTOG)
ules with concurrent chemotherapy for limited-stage small- Protocol 9712. Proc ASCO 22:631
cell lung cancer. J Clin Oncol 16(11):3528–3536 Komaki R, Scott C, Ettinger D, Lee JS, Fossella FV, Curran W
Christian JA, Bedford JL, Webb S, Brada M (2007) et al (9204) Randomized study of chemotherapy/radiation
Comparison of inverse-planned three-dimensional confor- therapy combinations for favorable patients with locally
mal radiotherapy and intensity-modulated radiotherapy for advanced inoperable nonsmall cell lung cancer: Radiation
non-small-cell lung cancer. Int J Radiat Oncol Biol Phys Therapy Oncology Group (RTOG). Int J Radiat Oncol Biol
67(3):735–741 Phys 38:149–155
Cox JD (1985) Large dose fractionation (hypofractionation). Lee JS, Scott CB, Komaki R, Ettinger DS, Sause WT (2002)
Cancer 55:2105–2111 Impact of institutional experience on survival outcome of
Cox JD, Gingerelli F, Ream NW, Maier JG (1972) Total patients undergoing combined chemoradiation therapy for
pulmonary irradiation for metastases from testicular carci- inoperable non-small-cell lung cancer. Int J Radiat Oncol
noma. Radiology 105:163–167 Biol Phys 52(2):362–370
Diener-West M, Pajak TF, Bauer M, Cox JD (1991) Liao Z, Komaki R, Stevens C (2002) Twice daily irradiation
Randomized dose searching phase ILE/II trials of fractiona- increases locoregional control in patients with medically
tion in radiation therapy for cancer. J Natl Cancer Instit inoperable or surgically unresectable stage II–IIIB non-small-
83:1065–1071 cell lung cancer. Int J Radiat Oncol Biol Phys 53(3):558–565
Din OS, Lester J, Cameron A, Ironside J, Gee A, Falk S et al Liao ZX, Komaki RR, Thames HD Jr, Liu HH, Tucker SL,
(2008) Routine use of continuous, hyperfractionated, Mohan R et al (2010) Influence of technologic advances on
accelerated radiotherapy for non-small-cell lung cancer: a outcomes in patients with unresectable, locally advanced
five-center experience. Int J Radiat Oncol Biol Phys 72(3): non-small-cell lung cancer receiving concomitant chemo-
716–722 radiotherapy. Int J Radiat Oncol Biol Phys 76(3):775–781
Fu KK, Pajak TF, Trotti A, Jones CU, Spencer SA, Phillips TL Liu HH, Balter P, Tutt T, Choi B, Zhang J, Wang C et al (2007)
et al (2000) A Radiation Therapy Oncology Group (RTOG) Assessing respiration-induced tumor motion and internal
phase III randomized study to compare hyperfractionation target volume using four-dimensional computed tomog-
and two variants of accelerated fractionation to standard raphy for radiotherapy of lung cancer. Int J Radiat Oncol
fractionation radiotherapy for head and neck squamous cell Biol Phys 68(2):531–540
Radiation Time, Dose, and Fractionation in the Treatment of Lung Cancer 141

McCracken JD, Janaki LM, Crowley JJ (1990) Concurrent concomitant cisplatin and radiotherapy on inoperable non-
chemotherapy/radiotherapy for limited small-cell lung carci- small cell lung cancer. N Engl J Med 326:524–530
noma: a Southwest Oncology Group Study. J Clin Oncol Sejpal S, Komaki R, Tsao A, Chang JY, Liao Z, Wei X et al
8:892–898 (2011) Early findings on toxicity of proton beam therapy
Michalski D, Sontag M, Li F, de Andrade RS, Uslene I, with concurrent chemotherapy for nonsmall cell lung
Brandner ED et al (2008) Four-dimensional computed cancer. Cancer
tomography-based interfractional reproducibility study of Spiro SG, James LE, Rudd RM, Trask CW, Tobias JS, Snee M
lung tumor intrafractional motion. Int J Radiat Oncol Biol et al (2006) Early compared with late radiotherapy in
Phys 71(3):714–724 combined modality treatment for limited disease small-cell
Murray N, Coy P, Pater JL, Hodson I, Arnold A, Zee BC et al lung cancer: a London Lung Cancer Group multicenter
(1993) Importance of timing for thoracic irradiation in the randomized clinical trial and meta-analysis. J Clin Oncol
combined modality treatment of limited-stage small-cell 24(24):3823–3830
lung cancer. The National Cancer Institute of Canada Thames HD, Peters LJ, Withers HR, Fletcher GH (1983)
Clinical Trials Group. J Clin Oncol 11(2):336–344 Accelerated fractionation versus hyperfractionation: ratio-
Perez CA, Stanley K, Rubin P, Kramer S, Brady L, Perez- nales for several treatments per day. Int J Radiat Oncol Biol
Tamayo R et al (1988) A prospective randomized study of Phys 9:127–138
various irradiation doses and fractionation schedules in Timmerman R, Papiez L, McGarry R, Likes L, DesRosiers C,
the treatment of inoperable non-small cell carcinoma of Frost S et al (2003) Extracranial stereotactic radioablation:
the lung. Preliminary report by the Radiation Therapy results of a phase I study in medically inoperable stage I
Oncology Group. Cancer 45:2744–2753 non-small cell lung cancer. Chest 124(5):1946–1955
Pignon JP, Arriagada R, Ihde DC, Johnson DH, Perry MC, Timmerman R, McGarry R, Yiannoutsos C, Papiez L, Tudor K,
Souhami RL et al (1992) A meta-analysis of thoracic DeLuca J et al (2006) Excessive toxicity when treating
radiotherapy for small-cell lung cancer. N Engl J Med central tumors in a phase II study of stereotactic body
327(23):1618–1624 radiation therapy for medically inoperable early-stage lung
Pijls-Johannesma M, De Ruysscher D, Vansteenkiste J, Kester cancer. J Clin Oncol 24(30):4833–4839
A, Rutten I, Lambin P (2007) Timing of chest radiotherapy Timmerman R, Paulus R, Galvin J, Michalski J, Straube W,
in patients with limited stage small cell lung cancer: a Bradley J et al (2010) Stereotactic body radiation therapy
systematic review and meta-analysis of randomised con- for inoperable early stage lung cancer. JAMA 303(11):
trolled trials. Cancer Treat Rev 33(5):461–473 1070–1076
Qiao X, Tullgren O, Ingmar L, Sirzen F, Lewensohn R (2003) Turrisi AT III, Kim K, Blum R, Sause WT, Livingston RB,
The role of radiotherapy in treatment of stage I non-small Komaki R et al (1999) Twice-daily compared with once-
cell lung cancer. Lung Cancer 41:1–11 daily thoracic radiotherapy in limited small-cell lung cancer
Rosenzweig KE, Sim SE, Mychalczak B, Braban LE, Schindel- treated concurrently with cisplatin and etoposide. N Engl J
heim R, Leibel SA (2001) Elective nodal irradiation in the Med 340(4):265–271
treatment of non-small-cell lung cancer with three-dimen- Uematsu M, Shioda A, Suda A et al (2001) Computed
sional conformal radiation therapy. Int J Radiat Oncol Biol tomography-guided frameless stereotactic radiotherapy for
Phys 50(3):681–685 stage I non-small-cell lung cancer: a 5-year experience. Int J
Rosenzweig KE, Sura S, Jackson A, Yorke E (2007) Involved- Radiat Oncol Biol Phys 51(3):666–670
field radiation therapy for inoperable non small-cell lung Warde P, Payne D (1992) Does thoracic irradiation improve
cancer. J Clin Oncol 25(35):5557–5561 survival and local control in limited-stage small-cell
Saunders MI (2000) The implications of the CHART trial for carcinoma of the lung? A meta-analysis. J Clin Oncol 10:
the treatment of non-small cell lung cancer. Lung Cancer 890–895
2:177–178 Yuan S, Sun X, Li M, Yu J, Ren R, Yu Y et al (2007) A
Saunders MI, Dische S, Barret S, Harvey A, Gibson D, Parmar randomized study of involved-field irradiation versus
M (1997) Continuous hyperfractionated accelerated radio- elective nodal irradiation in combination with concurrent
therapy versus conventional radiotherapy in small-cell lung chemotherapy for inoperable stage III nonsmall cell lung
cancer: a randomized multicentre trial. Lancet 350:161–165 cancer. Am J Clin Oncol 30(3):239–244
Sause WT, Scott C, Taylor S, Johnson D, Livingston R, Zhang X, Li Y, Pan X, Xiaoqiang L, Mohan R, Komaki R
Komaki R et al (1995) Radiation Therapy Oncology Group et al (2010) Intensity-modulated proton therapy reduces
(RTOG) 88-08 and Eastern Cooperative Oncology Group the dose to normal tissue compared with intensity-
(ECOG) 4588: Preliminary results of a phase III trial in modulated radiation therapy or passive scattering proton
regionally advanced, unresectable non-small cell lung therapy and enables individualized radical radiotherapy
cancer. J Natl Cancer Inst 87(3):198–205 for extensive stage IIIB non-small-cell lung cancer: a
Schaake-Koning C, van den Bogaert W, Dalesio O, Festen J, virtual clinical study. Int J Radiat Oncol Biol Phys 77(2):
Hoogenhourt J, van Houtte P et al (1992) Effects of 357–366
3D Radiation Treatment Planning
and Execution
Mary K. Martel

Contents Abstract
Technological advances have become commer-
1 Introduction.............................................................. 143 cially available and widely implemented. In par-
2 Treatment Planning Process .................................. 144 ticular, 3D conformal therapy has become the first
2.1 Introduction................................................................ 144 step in improving the targeting of dose to the
2.2 Immobilization and Simulation................................. 145 tumor while sparing dose to normal tissue. The
2.3 Planning Target Volume ........................................... 145
treatment planning process including beam design,
3 Radiation Beam Design and Delivery ................... 148 treatment planning objectives, and dose calculation
3.1 Standard Beam Arrangements................................... 148 issues for 3D radiation treatment planning will be
3.2 3D Conformal Treatment Techniques ...................... 150
reviewed. Topics such as target volume definition,
4 Normal Tissue Tolerance and Treatment use of imaging, set-up uncertainties, respiration
Planning Objectives................................................. 151
control, and normal tissue tolerance are briefly
5 Dose Calculation Issues........................................... 153 introduced.
5.1 Effects of Lung Density ............................................ 153
5.2 Calculation Algorithms ............................................. 154
6 Summary................................................................... 155
References.......................................................................... 155 1 Introduction

The goal of radiotherapy is to deliver therapeutic dose


in a precise and accurate manner to the target volume
while minimizing dose to surrounding normal tissue.
Advancement in technology over the past several
decades has brought highly developed means to reach
this objective. Planning and delivery of radiation
therapy has evolved to a multi-step process which is
individualized for each patient. This process includes
anatomy definition (including tumor and important
normal structures), radiation beam design, delivery of
the treatment plan, and verification of delivery. The
complexity of the treatment process depends on many
M. K. Martel (&)
factors; of paramount importance is the level of dose
Department of Radiation Physics,
Division of Radiation Oncology, prescription and whether the ultimate clinical intent is
University of Texas MD Anderson Cancer Center, curative.
1515 Holcombe BLVD, Houston, TX 77030, USA
e-mail: mmartel@mdanderson.org

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 143
DOI: 10.1007/174_2011_302, Ó Springer-Verlag Berlin Heidelberg 2011
144 M. K. Martel

For treatment of non-small cell lung cancer survival rate per day for treatment prolongation
(NSCLC), radiation therapy (RT) dose prescriptions beyond 6 weeks (Fowler and Chaell 2000). Accord-
with standard dose per fraction range from 60 to ingly, dose escalation schemes that limit overall
70 Gy at 1.8 to 2 Gy per fraction. However, it appears treatment time to 6 weeks or less provide a potential
from clinical data that 70 Gy may translate to a tumor ‘‘radiobiological’’ avenue to explore for improvement
control probability (or local progression-free survival) of outcome. Several trials are already under-
of approximately 30% (Martel et al. 1999; Hazuka way (Mehta et al. 2001; Belderbos et al. 2003;
et al. 1993). Supporting this outcome data are results Timmerman et al. 2003). The subject of time, dose
from other single institution trials (using standard and fractionation of radiation therapy is explored in
doses) which show overall survival rates of 33–43% depth (Cox) elsewhere in this book.
(Armstrong et al. 1995; Graham et al. 1995; Sibley Another major source of failure is geographic miss
et al. 1995). In addition, however, several of these of the tumor target volume by the planned radiation
trials, along with the multi-institutional RTOG 8301 fields. This is mainly due to effects related to the
altered fractionation trial (Cox et al. 1990), saw an anatomical site of the tumor in the lung, namely,
elevated incidence of high grade pneumonitis. These respiration effects causing tumor movement, and the
modest local control and survival rates coupled with uncertainties of the radiation dose calculation due to
undesired normal lung toxicity led to a rethinking of the lower density of the lung. Also as well, the
the radiotherapy treatment approach. Interest in RT treatment planning phase is highly dependent on how
dose escalation beyond 70 Gy launched a series of target volumes are determined for a given patient.
phase I trials aimed to determine the maximum tol- Inadequate tumor definition from imaging studies
erated dose (Robertson et al. 1997; Armstrong et al. leads to a target volume that does not cover the full
1997; Rosenzweig et al. 2000; Belderbos et al. 2003), extent of the disease, and geographic miss will occur.
with secondary endpoints to determine impact on The solutions to minimize geographic miss are tech-
local control and survival. However, given the dose- nological in nature and will be briefly discussed in
volume relationship for normal lung with toxicity, this chapter. The basic and advanced technical aspects
(Martel et al. 1994; Oetzel et al. 1995; Graham of treatment planning for radiation therapy will be
et al. 1999; Marks et al. 1997; Kwa et al. 1998; covered, which will serve as an introduction to later
Sepenwoolde et al. 2003) a novel dose escalation chapters that describe target volume definition, nor-
scheme (Ten Haken et al. 1993) was designed so that mal tissue toxicity, respiration control, and advanced
the prescribed dose would depend on the amount of delivery techniques such as stereotactic radiotherapy
normal lung volume irradiated, rather than escalate in and intensity modulated radiation therapy in greater
the standard fashion. A normal tissue complication detail.
probability (NTCP) model for the calculation of risk
of pneumonitis was used to set the dose levels so that,
as the dose was escalated, the risk of toxicity 2 Treatment Planning Process
increased in a predictable manner. Using this design,
doses were escalated well above standard doses, 2.1 Introduction
achieving 84–102.9 Gy for many patients (Kong et al.
2006) without the development of pneumonitis. For In simple terms, radiotherapy treatment planning can
the subgroup of patients receiving doses greater than be defined as the process of arrangement of beams to
92.4 Gy (Narayan et al. 2004), survival rates irradiate a defined target volume to the prescribed
improved. However, local control remained prob- dose. The accuracy of beam targeting improved in the
lematic, with progression occurring for many of the 1980s with the advent of computerized image-based
patients. treatment planning which allowed the widespread use
It has been hypothesized that one source of failure of dose calculations in three dimensions (3D) based
of high doses to control all tumors is due to the on patient-specific 3D anatomy. For the anatomical
possible accelerated repopulation late in the treatment site of the thorax, treatment planning is complicated
course of 8–10 weeks needed to deliver doses in by the number of normal organs (spinal cord, normal
excess of 80 Gy. Estimates predict a 1.6% loss of lung, esophagus and heart) located close to the tumor,
3D Radiation Treatment Planning and Execution 145

which have limited tolerance to radiation. However, approximate the location of soft tissue target volumes.
‘‘3D’’ technology has allowed reduced irradiation of Computed tomography (CT) scan information sup-
normal tissues by design of field shapes with the plements, and, now more commonly, replaces the
‘‘beam’s eye view’’ and arrangement of multiple non- simulator X-rays for soft tissue volume delineation.
coplanar, non-axial beam angles with 3D visualiza- Axial images are acquired with thin slices (3–5 mm)
tion tools (McShan et al. 1990). This allows dose to through the target area and adjacent normal struc-
‘‘conform’’ to the tumor/target volume while maxi- tures, usually from vertebral bodies C4 to L1 at a
mizing sparing of dose to surrounding normal tissue; minimum, and to include the entire volume of both
this technique is called ‘‘conformal’’ therapy. Dose lungs. A coordinate system must be established
distributions calculated in 3D can be evaluated between the imaging studies and the treatment
throughout the 3D patient volume, allowing for machine. This is accomplished through the use of an
detailed analysis to facilitate achievement of the alignment system common to the simulator, CT
optimal plan. The intricacies of the treatment plan- scanner, and treatment rooms. Wall mounted lasers
ning process for both standard and conformal radio- project lines in three planes (axial, sagittal and coro-
therapy techniques for lung cancer will be reviewed nal) and intersect at the isocenter, defined as the focal
here. In addition, Senan et al. (2004) have an excel- point of the treatment linear accelerator’s rotation at a
lent review of literature-based recommendations for point in space. In an X-ray simulator or CT simulator
treatment planning for the lung. (a CT scanner with a laser system and localization
software), the patient is aligned so that the approxi-
mate center of the tumor is positioned at the isocenter.
2.2 Immobilization and Simulation An example of the placement of the isocenter during
the CT simulation process is given in Fig. 1. The field
3D planning begins with the acquisition of an imaging center and border can be display by the simulation
volume data set with the patient in the treatment software and the isocenter can be placed via software
position. First, the patient is placed on a support table tools to the center of the tumor, using the coronal
in a position that can be easily reproduced during (left) and axial (right) reconstructed CT images as
treatment setup. For patients with lung cancer, opti- guidance. Since the lasers are aligned to point to the
mally the arms are positioned above their head so as isocenter, the intersection of the lasers with the
not to restrict selection of beam angles and prevent patient’s skin surface is then marked in the simulator.
treatment through the arms. A positioning (otherwise These reference marks are used to re-align the
known as ‘‘immobilization’’) device is used to help patients at the time of daily treatment at the linear
duplicate the same position for each day of treatment. accelerator.
It is now common to make custom devices to fit
individual patients. Custom foam cradles are used for
immobilization of thorax region. A foam mixture fills 2.3 Planning Target Volume
the space between a styrofoam form and the patient,
forming to the body shape, which is then attached 2.3.1 ICRU Guidelines
with pegs to the treatment table. Well made immo- The imaging studies are used to construct a target
bilization devices will reduce the magnitude of daily volume, the first crucial step in the planning process.
set-up uncertainty. To promote systematic target volume definition, the
Localization of patient anatomy is performed using International Commission on Radiation Units and
imaging studies. The simplest method is the use of a Measurements (ICRU) has published nomenclature
machine called a simulator that has the same geo- and guidelines (ICRU 1993, 1999). For target volume
metrical features of a linear accelerator (identical delineation, several concentric volumes are described.
isocentric gantry design and position of treatment b), First, the extant of malignant cells visible on imaging
but has a diagnostic X-ray generator in the head of the studies, including any involved nodes, is called the
machine. It produces radiographs in two dimensional gross tumor volume (GTV). Next, a margin around
planes that visualize the intended anatomic area of the GTV is added to account for potential local–
treatment, but often using bony landmarks to regional subclinical extension, and is called the
146 M. K. Martel

Fig. 1 Coronal digital reconstructed radiograph (DRR) (left) showing placement of an anterior field isocenter; and an axial CT
slice with placement of the isocenter at depth in the patient

clinical target volume (CTV). The GTV and the CTV when followed, resulted in a reduction in the variation
are based solely on anatomic and biological consid- to 7–22% (average 13%). Reduction of the contouring
erations. The final volume is the planning target variation on CT is important since studies often relate
volume (PTV). This volumetric expansion accounts clinical outcome to tumor size or volume. The
for the uncertainties of the geographic position of the recommended guidelines are given below, which are
CTV from day-to-day. Specifically, a margin is added adapted from the procedure for the measurement of
to compensate for physiologic changes in the size, the volume of the primary tumor and involved lymph
shape, and position of internal anatomy. Additional nodes from the TROG 99-05 study (Bowden et al.
margins are added to account for patient movement 2002): Preparation: Volume measurements will be
(e.g., breathing) and differences in patient positioning based on planning CT images, which should be con-
from day to day (set-up uncertainty). The construction trast-enhanced. If the planning image does not have
of the PTV is discussed below. In addition, Armstrong contrast, a recent (within 2 weeks of planning) diag-
(1998), and Senan, Kepka and Videtic in later chap- nostic contrast-enhanced CT scan should be available
ters discuss target volume definition. for viewing alongside the planning CT scan. The
planning CT scan should include all known tumor and
2.3.2 Use of CT for Gross Tumor Volume all enlarged intrathoracic lymph nodes. Steps: (1)
Delineation Identification of tumor and nodes: The contour should
CT imaging is the most common modality used for therefore closely hug the surface of the tumor and
treatment planning. However, distinguishing tumor should not include a margin for suspected or micro-
from surrounding normal lung and soft tissue is often scopic spread. Opacities thought unlikely to represent
not straightforward, even with the use of contrast, and tumor, but that a prudent radiation oncologist might
there can be large variations in contoured volumes include in the CTV because of lack of absolute cer-
among clinicians and institutions (Senan et al. 1999; tainty, should be excluded. The tumor, plus all hilar
Bowden et al. 2002). Bowden et al. (2002) found that and mediastinal nodes with a diameter [1 cm, should
despite input from radiologists, significant variation be identified and outlined with a fine-tip felt pen by a
up to 42% (on average 20%) occurred in the delin- diagnostic radiologist on the hard copy. (2) Initially
eation of the 3D gross tumor volumes of NSCLC the volume is contoured using mediastinal window
among oncologists. The authors propose standardi- (MW) settings (width 400 HU and level +20 HU).
zation of the approach and give guidelines, which Because the density scale on commercial planning
3D Radiation Treatment Planning and Execution 147

systems does not always correspond with Hounsfield be aligned with the CT set by transforming the PET
units, it is recommended that the window settings be coordinate system to match the planning CT. Several
standardized for each individual department and that types of algorithms exist to achieve this transforma-
the same settings be used on every occasion. It is tion (Pelizzari 1998). For example, one of the sim-
suggested that a diagnostic radiologist be asked to plest techniques is to identify anatomical landmarks
establish which settings most closely correspond with in each dataset and ‘‘tie’’ the two image sets together.
the range of Hounsfield units for both MWs and lung A more complex registration method uses the entire
window (LW), as described in the report by (Harris volume of image data (i.e., intensities of the image
et al. 1993). For disease involving the mediastinum, voxels) for matching of ‘‘mutual information’’. Once
the tumor/node edge should be defined by the inter- the different image series are registered, image fusion
face between the tumor/node and fat or contrast- software is used to display the two modalities
enhanced vessel using MW settings. (3) In practice, it simultaneously.
is easiest to determine the tumor volume using the
MW settings and then to enlarge this volume as 2.3.4 Microscopic Margin
required after changing to the LW settings. The LW Generally, the size of margin added to the GTV to
should most closely correspond with a level of -750 account for microscopic extent (ME) has been
HU and a window width of 850 HU. With these set- somewhat arbitrary (i.e., 5 mm), or not used at all.
tings, the volume can only be contoured at the lung/ However, Giraud et al. (2000) examined NSCLC
tumor interface, because all mediastinal definition is surgical specimens with adenocarcinoma (ADC) and
lost. The maximal cross-sectional dimension of the squamous cell carcinoma (SCC) histology. The mean
tumor should be measured and recorded using the LW value of ME was 2.69 mm for ADC and 1.48 mm for
window image. Special situations (1) Spicules: Only SCC. The usual 5 mm margin covers 80% of the ME
the solid portion of the tumor should be contoured. for ADC and 91% for SCC. To have 95% confidence
Fine spicules radiating into the surrounding lung that all tumor is included in the clinical target volume,
should not be included, because the interpretation of a margin of 8 and 6 mm must be chosen for ADC and
their size and significance varies considerably among SCC, respectively.
observers. (2) Cavitating tumor: If the tumor is cav-
itating, its volume will be taken to be that volume if 2.3.5 Setup Uncertainties
no cavitation were present. (3) Atelectasis: Patients
with adjoining atelectasis represent a special case. 2.3.5.1 Sources of Error
Sometimes the radiologist is able to distinguish ate- Patient orientation at treatment may be different from
lectatic lung from tumor, especially if liver window the planned position. This is due in large part to
settings are used (window width 150 HU, level 50 random variation, but some systematic effects are
HU). present. Some sources of error are given below. For
one, the location of the lasers used to indicate the
2.3.3 Addition of PET Scans isocenter may differ between the simulation and
18-FDG-positron emission tomography (PET) has treatment room. Also, the patient may not be marked
had a large impact on the delineation of the gross on the setup points in an exact manner in the CT
tumor volume for lung cancer because it images simulation room. The patient may be imaged on the
metabolically active tumor cells. In particular, PET treatment machine before radiation is applied, which
has several advantages over CT in distinguishing quantifies setup error, but there are limitations in the
tumor from collapsed lung or mediastinal structures, ability to visually reading the portal image, as
and benign from malignant lymph node enlargement. opposed to utilizing computer-aided graphical align-
A detailed discussion on the use of PET in lung ment tools. Also, based on the portal image, it is
cancer is given in a later chapter (MacManus). The common to correct the position of the patient only
PET (or other image datasets, such as MRI) infor- when the needed shift exceeds 5 mm or greater.
mation needs to be correlated to CT scans through the Further, there may be limitations in the ability to
use of image registration software. Image data in make the proper shift in patient position. Finally, the
the PET transmission/emission or PET/CT dataset can patient may move on the table after imaging but
148 M. K. Martel

before treatment commences. Studies to measure available that will contour structures having the same
patient setup errors should be carried out to estimate density, allowing rapid definition of an entire 3D
the margin to be used for the planning target volume region. For example, lungs have one-third of the
definition. Study results will be dependent on the density of soft tissue and can be easily differentiated
immobilization, simulation and treatment techniques from surrounding tissue. Surfaces for each structure
used at each individual institution. Portal imaging are generated from the segmented contours, and can
everyday (instead of the current once-per-week) will be viewed in any plane that is generated through the
certainly decrease the setup uncertainty. An alternate surface. In Fig. 2, the planning target volume in green
method would be to image every day during the first and the spinal cord volume in yellow are displayed as
week of treatment to determine an individualized overlays on several reconstructed CT plans, such as
margin for use during the remainder of the treatment. the axial, sagittal and coronal planes in the left panels.
However, it is clear that regular observation and This type of display is useful during treatment beam
correction for patient setup is a necessity. design.
The next step in the planning process is the design
2.3.5.2 Accounting for Respiratory Motion of radiation beam field or aperture. In the treatment of
Tumor motion due to respiration must be included in lung cancer, relatively simple beam arrangements
the planning target volume definition, and can be have been traditionally used. This is due in large part
determined at the time of imaging. Simulator X-ray to the prophylactic treatment of hilar, mediastinal and
films or planning CTs represent a ‘‘snapshot’’ of a in some cases, supraclavicular lymph nodes which
point in time during the respiration cycle, which may may be at risk for harboring microscopic disease,
not be at the same point under treatment conditions. which is called ‘‘elective’’ nodal irradiation (ENI).
The use of CT simulators is now common place, and Though the ENI volume is determined by anatomical
CT images may be acquired during different phases of landmarks located on simulator X-ray images or
the respiratory cycle (4D-CT). Incorporation of a reconstructed coronal CT planes, it is rarely (if ever)
margin for motion will increase the planning target contoured by the radiation oncologist as a separate
volume, and consequently, increase the amount of structure. It is common to treat the ‘‘approximated’’
normal lung that is irradiated. Alternately, respiration ENI volume, along with the contoured primary GTV/
can be suspended during the planning CT and treat- PTV, in large fields aimed from the anterior and
ment, as described by Wong et al. (1999), through the posterior (AP/PA) direction of the patient in parallel-
use of a device called active breathing control (ABC) opposed fashion (see 2). Since the spinal cord is
or through respiratory gated therapy (Kubo et al. irradiated by the AP/PA fields, this beam arrangement
2000), where radiation is delivered only at a certain can only be used until the tolerance dose of the cord is
phase of the respiration cycle when the target is in a reached, generally 45–50 Gy at 1.8–2 Gy/fraction.
known position. Respiration control and 4D-CT is Fields are then arranged ‘‘off-cord’’ to treat only the
discussed in detail in a later chapter (Senan). primary PTV. An example design of off-cord fields is
shown in Fig. 3. Here, beam apertures are planned
using 3D treatment planning software, called ‘‘virtual
3 Radiation Beam Design simulation’’. Initially the radiation field borders are
and Delivery set in a rectangular fashion. Then, beam directions are
selected by the use of the beam’s eye view (BEV)
3.1 Standard Beam Arrangements tool. Target and normal structures are viewed from
different directions in planes perpendicular to the
After imaging data is complete for a given patient, the beam’s central axis using BEV (see Figs. 2 and 3).
target volumes and normal anatomic structures must Structures are distinguished from each other by use of
be defined. Each structure is circled or contoured on different colors. In Fig. 3, the beam direction is
individual axial images, using image display work- angled away from the normal structure to separate the
stations. The contouring process segments the image PTV structure from the spinal cord. The beam shape
data into separate structures, each uniquely identified. is then modified by designing a block that will
Semi-automated and automated algorithms are allow full dose to the PTV but minimize dose to
3D Radiation Treatment Planning and Execution 149

Fig. 2 Gross tumor volume


(GTV) contours shown in
green and spinal cord
contours in yellow are
overlaid on the coronal DRR
(upper right), and on axial,
sagittal and coronal CT slices.
The projection of the field
borders on the patient is
shown in yellow in each panel

Fig. 3 To avoid delivering dose to the spinal cord (yellow ‘‘separation’’ of the cord from the tumor volume (outlined in
contours) with the anterior field (left), the head of the machine blue) (middle). Blocks to shield normal tissue are then drawn
must be rotated until the virtual simulation software shows (outside blue line) (right)

surrounding normal tissue. The block shape is repre- The MLC consists of a number of small leaves that
sented by the outermost shape in the BEV in Fig. 3. move independent of each other to form the planned
Blocks consist of heavy metallic material mounted on shape (displayed in the BEV in Fig. 4).
a tray which is placed in the head of the machine, or a Once beam angles and shapes are designed, dose
block substitute called a multileaf collimator (MLC). calculations are performed. Since it is not possible or
150 M. K. Martel

Fig. 4 Beam’s eye view


planning with non-coplanar
and non-axial beams to avoid
normal structures for an upper
lobe tumor (University of
Michigan UMPlan)

practical to measure a 3D dose distribution for each in the field angles after evaluation of the dose
patient situation, a general dose calculation system distribution.
must be used to ‘‘predict’’ the dose in the patient.
These calculations incorporate basic data that char-
acterize the radiation beam energy and geometry, 3.2 3D Conformal Treatment Techniques
such as depth dose curves and isodose information for
standard field sizes. The deposition of dose from The fundamental rule of treatment planning is the use
photon irradiation results from the generation of of multiple beams to concentrate the high isodose
secondary electrons. In the case of photon energies in region at the isocenter and in the PTV. Two opposed
the therapeutic range, electrons are primarily set in beams, such as the AP/PA fields described above, will
forward motion by Compton interactions (in which produce a more uniform dose distribution throughout
energy is both absorbed and scattered), which then the volume when compared to the use of a single beam.
penetrate deeper into tissue. Energy is deposited into However, when more than two beams are used, the
tissue as these electrons slow down. Computerized dose is further concentrated in the PTV and dose to
algorithms have been developed to combine the dose normal tissues can be further reduced. An ideal treat-
distributions generated by combinations of beams, ment plan is both conformal (high dose wraps closely
using individual patient information such as depth of around the PTV with rapid falloff to low doses) and
the point of calculation, external body contour, and homogeneous (± 5% variability of dose within the
various densities of anatomical structures. Dose dis- PTV). Two example cases of conformal beam
tributions are displayed with concentric curves for arrangements are given below. Shown in Fig. 4 is a
chosen dose levels (isodoses) which are displayed as tumor of the upper lobe. Normal tissue structures such
overlays on anatomic structures. These curves are as lungs, heart, esophagus and spinal cord are con-
normalized to a reference dose, either at isocenter or toured on CT, and are displayed as solid surfaces in 3D
to the lowest isodose curve that encompasses the in a variety of BEV displays. The PTV, in red, is tar-
PTV. Ideally, the 95% isodose curve will cover the geted by beams directed from five different machine
planning target volume, or adjustments will be made gantry angles, and several different couch angles.
3D Radiation Treatment Planning and Execution 151

shown in the lower left panel. The dose-volume


histograms are shown in Fig. 7.
If the treatment plan does not meet the given
dose-volume objectives, beam arrangements or other
parameters are adjusted. This can include a change of
beam energy, beam angle, or adjustment of the beam
intensity. For ‘‘forward-planned’’ conformal therapy,
these adjustments are carried out manually, with chan-
ges made in an iterative fashion by the treatment planner.
Normally the beam intensity is uniform across the beam
width and length. The simplest modification of the
intensity is a wedged shape filter placed in the machine
head. A more complex method is to break the field
aperture into segments with varying beam-on times.
Currently the most intricate form of intensity modulation
achieves a checkerboard pattern with each square of a
Fig. 5 Dose-volume histograms for treatment plan shown in varying intensity. The delivery of this type of pattern is
Fig. 4 with a compensator or with a device called a multileaf
collimator, which can move under computer control to
These angles were chosen so that each normal structure shape segments of the field to deliver the intensity pat-
is irradiated by only several, but not all, of the beams. tern. This is called intensity modulated radiation therapy
For example, the spinal cord, in green, is contained (IMRT) and with this technique, there is potentially a
within two of the 6 BEV fields. If the dose distribution high degree of control over the shaping of the dose dis-
for the PTV is not homogeneous, segments of fields tribution. IMRT for treatment of lung cancer is described
may be placed to ‘‘boost’’ the dose; one such segment is in detail in a later chapter (Grills et al. 2003).
shown in the lower right of Fig. 4. Once the final beams are designed, X-ray images in
The treatment plan can be evaluated as to whether the form of digitally reconstructed radiographs (DRRs)
it meets objectives of PTV coverage and normal tis- are generated from the treatment planning CT to
sue avoidance. A set of criteria for normal tissue enhance the bony anatomy with high contrast, and are
tolerances (discussed in the next section) must be in the beam’s eye view plane. An example of a DRR of
given to guide the treatment planner. Dose distribu- the lung is shown in Fig. 3. DRRs are used to compare
tions for 3D volumes can be displayed and analyzed to portal images taken before treatment, which are
graphically with dose-volume histograms (DVH), either films placed in the beam exiting the patient or
generated for each structure. The cumulative form of with an electronic portal device. Verification of the
the DVH is a plot of the volume of a given structure radiation beam placement vis-à-vis the patient is car-
receiving a certain dose or higher as a function of ried out pre-treatment so that patient position can be
dose. DVHs for the beam arrangement in Fig. 4 are adjusted accordingly. Beam delivery is carried out with
displayed in Fig. 5. The dose-volume histogram for use of beam modifiers such as blocks, multileaf colli-
normal lung is the addition of the dose distributions of mators, wedges, compensators, or IMRT.
both lungs but minus the dose distribution in the
GTV. The GTV is selected instead of the PTV, since
the PTV contains normal lung receiving high dose 4 Normal Tissue Tolerance
which influences the normal tissue toxicity rate. and Treatment Planning Objectives
The second case is shown in Fig. 6. The tumor is
centrally located and in the lower lobe. Though the Three dimensional conformal therapy is now a mature
PTV is located near the spinal cord, dose to the cord is technology in widespread use. However, it is still
kept below tolerance by shielding the structure in two difficult to design the ‘‘best’’ plan, defined as a bal-
of the four fields. However, the PTV is also blocked, ance of achieving high dose delivery to the tumor
and the dose is boosted by adding a field segment, with a low rate of normal tissue toxicity. A set of criteria
152 M. K. Martel

Fig. 6 Beam’s eye view


planning with non-coplanar
and non-axial beams to avoid
normal structures for an lower
lobe tumor (University of
Michigan UMPlan)

for normal tissue tolerances should be established from


published studies and adapted for local clinical use. A
good starting point is the report by a National Cancer
Institute-sponsored task force which carried out an
extensive literature search and presented updated
information on tolerance of normal tissues, with
emphasis on partial volume effects (Emami et al. 1991).
For uniform irradiation of normal lung, tolerance
doses for a 5% chance of pneumonitis occurring within
5 years for uniform irradiation of 1/3 of the lung was
45 Gy, 2/3 was 30 Gy and whole lung was 17.5 Gy.
For the esophagus, the corresponding doses are: 60 Gy
(1/3), 58 Gy (2/3) and 55 Gy (whole) for an endpoint
of clinical stricture/perforation. For the heart: 60 Gy
(1/3), 45 Gy (2/3) and 40 Gy (whole) for the endpoint of
pericarditis. The 50% chance of a complication occur-
ring in 5 years was also given for each organ. These Fig. 7 Dose-volume histograms for treatment plan shown in
Fig. 6
tolerance data show that the complication probability
may be a function of irradiated volume and dose.
The Emami tolerance data summary was one of the doses given were based on limited volumetric dose
early efforts towards the use of objective criteria in data publications and on ‘‘guesstimates’’ based on
evaluating treatment plans. However, the tolerance clinical experience. As 3D dose distributions for
3D Radiation Treatment Planning and Execution 153

Hayman et al. 2001) at the University of Michigan.


Because of the observed dose-volume relationship
for normal lung with toxicity, the prescribed dose
depended on the volume of lung irradiated by the
plan, rather than escalate in the standard fashion
with all patients regardless of the amount of lung
irradiated receiving the same level of dose. A normal
tissue complication probability (NTCP) model was
used to set the dose levels so that, as the dose was
escalated, the risk of pneumonitis increased in a
predictable manner. The Netherlands Cancer Insti-
tute (Belderbos et al. 2003) has a similar approach
but use mean lung dose to stratify patients into dose
groups. The Radiation Therapy Oncology Group’s
(RTOG) trial (RTOG 1993) has three levels of
stratification according to V20. The use of 3D con-
formal therapy is a requirement for these studies.
Dose escalation will be discussed in a later chapter
Fig. 8 The incidence of radiation pneumonitis as a function of
the mean normalized total dose (NTDmean), representing mean (Rosenzweig).
lung dose (Kwa et al. 1998)

5 Dose Calculation Issues


normal lung became available, dosimetric parameters
could be correlated with complication data. Martel 5.1 Effects of Lung Density
et al. (1994), Oetzel et al. (1995), Marks et al. (1997),
Graham et al. (1999) related dose-volume metrics to It is generally recommended to use density correc-
the risk of developing pneumonitis. Kwa et al. (1998) tions and low energy photon beam for treatment
found a relationship between the incidence of radia- planning of lung cancer. When density is not taken
tion pneumonitis and the mean lung dose in an anal- into account in computer calculations, lungs are
ysis of pooled data of 540 patients from five assigned a density of one, which is equivalent to
institutions of the previously listed studies. Increasing water, instead of approximately 0.2–0.4 that is reality.
mean lung dose correlated well with increasing This means that the attenuation of photons per unit
pneumonitis rate. Figure 8 illustrates this relationship length is lower in low density lung tissue compared
and can be used to reliably predict the risk of pneu- with unit density water-equivalent tissue. When dose
monitis when mean lung dose is evaluated from a was measured in a benchmark test phantom to a point
treatment plan DVH. Each of these studies provides in between two lungs, there was increased dose ran-
dosimetric parameters that can be extracted from the ging from 5 to 14% relative to a phantom of unit
3D dose distribution to give the clinician a guide for density. The effect decreases as the photon energy
safe treatment. The Emami data have been recently increases. The use of high energy beams could be
updated by an AAPM/ASTRO/NCI group called used to minimize the dose correction discrepancies.
‘‘Quantitative Analyses of Normal Tissue Effects in However, studies (Mackie et al. 1985; Rice et al.
the Clinic’’ (QUANTEC) (Bentzen et al. 2010). 1988) have shown that higher energy beams tend to
Updated treatment related toxicity information is ‘‘spare’’ the surface of the tumor when traversing
given in detail in later chapters. through the lung. Too, higher energies will have an
Complication data such as those discussed above increased range of secondary electrons in lung tissue,
have helped in the design of dose escalation trials. which further spreads out the low isodoses relative to
As discussed in the introduction, one of the first a water-equivalent tissue (Ekstrand and Barnes 1990).
trials in the 3D treatment planning era used a novel When a clinically relevant phantom study was per-
dose escalation scheme (Ten Haken et al. 1993; formed (Klein et al. 1997), dose delivered to the PTV
154 M. K. Martel

Fig. 9 Isodose distributions in transverse and coronal views white, 20%. The colorwashes in red and blue represent the
through the dose specification point of a five-field plan of a GTV and the PTV, respectively. Note the difference in the
treatment of a right hilar NSCLC, computed using (left) the computations of the 95% isodose line in the PTV (De Jaeger
EPL algorithm and (right) the CS algorithm. The isodose levels et al. 2003)
displayed are: blue, 95%; pink, 90%; yellow, 80%; green, 50%;

with 6 MV was within 5% of predicted, but low by The limitation of these algorithms is that the increased
11% with use of 18 MV. lateral electron scatter in lung tissue is not accounted
for. Calculation algorithms have become more
sophisticated in the past decade and practical to use
5.2 Calculation Algorithms with the increase in computer calculation power. For
example, the convolution-superposition (CS) method
Current treatment-planning computer systems have the can predict the lack of lateral electron transport in the
capability of incorporating the effect of lower lung calculation. The effect of the more accurate CS algo-
density into the dose calculation, and there are several rithm versus the EPL algorithm is shown in Fig. 9
density-correction algorithms. However, because of (De Jaeger et al. 2003). The patient’s original plan
the variety of treatment situations for lung cancer, it is using EPL shows that the 95% isodose line is enclosing
difficult to take into account all effects, such the build- the PTV (left panels). Recalculation of the dose dis-
up and scattering of secondary electrons. Such effects tributions with the CS model shows that the 95% iso-
depend on, for example, how much lung is traversed, dose line constricts into the PTV, causing a reduction
beam energy, and beam field size. Correction-based of dose particularly in the region of the PTV that is
algorithms such as the equivalent-pathlength (EPL) embedded in lung, due to the penumbra broadening in
model, the generalized Batho and equivalent-tissue-air the low-density lung tissue. This effect is less at the
ratio (ETAR) methods are available commercially. mediastinal boundary with the PTV. Overall, the mean
3D Radiation Treatment Planning and Execution 155

lung dose as determined by the CS and EPL algorithms decades is to determine the impact of new technology
differed on average 17%, and the V20 differed on on treatment outcome.
average by 12% (De Jaeger et al. 2003).
Model-based calculation techniques, such as the
convolution-superposition and more recently, Monte References
Carlo methods, offers a physics-based approach found
to be more accurate than correction-based methods Armstrong JG (1998) Target volume definition for three-
for calculating the dose in inhomogeneous media. The dimensional conformal radiation therapy of lung cancer. Br
J Radiol 71:587–594
Monte Carlo method is the only method that explicitly
Armstrong JG, Zelefsky MJ, Leibel SA et al (1995) Strategy for
transports photons and electrons within a material and dose escalation using 3-dimensional conformal radiation
is therefore likely to provide more accurate results at therapy for lung cancer. Ann Oncol 6:693–697
material interfaces and within lower density material Armstrong J, Raben A, Zelefsky M et al (1997) Promising
survival with three-dimensional conformal radiation therapy
(Chetty et al. 2003). A wide range of experiments
for non-small cell lung cancer. Radioth Oncol 44:17–22
have been conducted in both unit density and low Belderbos JS, De Jaeger K, Heemsbergen WD et al (2003) First
density geometries to validate user-specific Monte results of a phase I/II dose escalation trial in non-small cell
Carlo codes developed for clinical treatment plan- lung cancer using three-dimensional conformal radiother-
apy. Radiother Oncol 66:119–126
ning. When Monte Carlo was used to recalculate
Bentzen SM, Constine LS, Deasy JO et al (2010) Quantitative
patient cases (Wang et al. 2002) the calculated dose analyses of normal tissue effects in the clinic (QUANTEC):
distributions were again characterized by reduced an introduction to the scientific issues. Int J Radiat Oncol
penetration and increased penumbra due to larger Biol Phys 76:S3–S9
Bowden P, Fisher R, Mac Manus M et al (2002) Measurement
secondary electron range in the low-density media,
of lung tumor volumes using three-dimensional computer
not as accurately accounted for in the pencil beam planning software. Int J Radiat Oncol Biol Phys 53:566–573
algorithm compared to Monte Carlo. It was concluded Chetty IJ, Charland PM, Tyagi N et al (2003) Photon beam
that it would be optimal to either use a Monte Carlo relative dose validation of the DPM Monte Carlo code in
lung-equivalent media. Med Phys 30:563–573
once fast algorithms are developed.
Cox JD, Azarnia N, Byhardt RW et al (1990) A randomized
phase I/II trial of hyperfractionated radiation therapy with
total doses of 60.0 Gy to 79.2 Gy: possible survival benefit
6 Summary with greater than or equal to 69.6 Gy in favorable patients
with Radiation Therapy Oncology Group stage III non-
Technological advances have become commercially small-cell lung carcinoma: report of Radiation Therapy
Oncology Group 83-11. J Clin Oncol 8:1543–1555
available in the past decade. In particular, 3D con- De Jaeger K, Hoogeman MS, Engelsman M et al (2003)
formal therapy has become the first step in improving Incorporating an improved dose-calculation algorithm in
the targeting of dose to the tumor while sparing dose conformal radiotherapy of lung cancer: re-evaluation of
to normal tissue, and has facilitated radiation dose dose in normal lung tissue. Radiother Oncol 69:691–710
Ekstrand KE, Barnes WH (1990) Pitfalls in the use of high
escalation. Though local control and survival has not energy X rays to treat tumors in the lung. Int J Radiat Oncol
yet dramatically improved with recent dose escalation Biol Phys 18:249–252
trials, this may possibly due to geographical misses Emami B, Lyman J, Brown A et al (1991) Tolerance of normal
because of poor target definition, movement of the tissue to therapeutic irradiation. Int J Radiat Oncol Biol
Phys 21:109–122
tumor due to respiration, and dose/fractionation lev- Fowler JF, Chaell R (2000) Non small cell lung tumors
els. Improved construction of the planning target repopulate rapidly during radiation therapy. Int J Radiat
volume is an important first step in improving the Oncol Biol Phys 46:516–517
treatment planning process. Further improvements Giraud P, Antoine M, Larrouy A (2000) Evaluation of
microscopic tumor extension in non-small-cell lung cancer
can be gained by sophisticated beam arrangement for three-dimensional conformal radiotherapy planning. Int
planning, made possible with intelligent choice of J Radiat Oncol Biol Phys 48:1015–1024
clinically relevant normal tissue tolerance criteria. Graham MV, Purdy JA, Emami B et al (1995) Preliminary
Finally, algorithms to account for the effects of lower results of a prospective trial using three dimensional
radiotherapy for lung cancer. Int J Radiat Oncol Biol Phys
lung density have become available and will facilitate 33:993–1000
the accurate and realistic calculation of dose to the Graham MV, Purdy JA, Emami BE et al (1999) Clinical dose
PTV and the lung. The next step in the coming volume histogram analysis for pneumonitis after 3D
156 M. K. Martel

treatment for non-small cell lung cancer (NSCLC). Int J Mehta M, Scrimger R, Mackie R et al (2001) A new approach
Radiat Oncol Biol Phys 45:323–329 to dose escalation in non-small-cell lung cancer. Int J Radiat
Grills IS, Yan D, Martinez AA et al (2003) Potential for Oncol Biol Phys 49:23–33
reduced toxicity and dose escalation in the treatment of Narayan S, Henning GT, Ten Haken RK et al (2004) Results
inoperable non-small-cell lung cancer: a comparison of following treatment to dose of 92.4 or 102.9 Gy on a phase I
intensity-modulated radiation therapy (IMRT), 3D confor- dose escalation study for non-small cell lung cancer. Lung
mal radiation, and elective nodal irradiation. Int J Radiat Cancer 44:79–88
Oncol Biol Phys 57:875–890 Oetzel D, Schraube P, Hensley F et al (1995) Estimation of
Harris KM, Adams H, Lloyd DCF et al (1993) The effect of pneumonitis risk in three-dimensional treatment planning
apparent size of simulated pulmonary nodules of using three using dose-volume histogram analysis. Int J Radiat Oncol
standard CT window settings. Clin Radiol 47:241–244 Biol Phys 33:455–460
Hayman JA, Martel MK, Ten Haken RK et al (2001) Dose Pelizzari CA (1998) Image processing in stereotactic planning:
escalation in non-small-cell lung cancer using three-dimen- volume visualization and image registration. Med Dosim
sional conformal radiation therapy: update of a phase I trial. 23:137–145
J Clin Oncol 19:127–136 Radiation Therapy Oncology Group RTOG 93-11 (1993) A
Hazuka MB, Turrisi AT 3rd, Lutz ST et al (1993) Results of phase I/II dose escalation study using three dimensional
high-dose thoracic irradiation incorporating beam’s eye conformal radiation therapy in patients with inoperable
view display in non-small cell lung cancer: a retrospective nonsmall cell lung cancer web page: www.rtog.org
multivariate analysis. Int J Radiat Oncol Biol Phys Rice RK, Mijnheer BJ, Chin LM (1988) Benchmark measure-
27:273–284 ments for lung dose corrections for X-ray beams. Int J
ICRU (1993) Prescribing, recording and reporting photon beam Radiat Oncol Biol Phys 15:399–409
therapy, Report 50, ICRU Press, Bethesda Robertson JM, Ten Haken RK, Hazuka MB et al (1997) Dose
ICRU (1999) Prescribing, recording and reporting photon beam escalation for non-small cell lung cancer using conformal
therapy (Su lement to ICRU Report 50) ICRU Press, radiation therapy. Int J Radiat Oncol Biol Phys
Bethesda 37:1079–1085
Klein EE, Morrison A, Purdy JA et al (1997) A volumetric Rosenzweig KE, Mychalczak B, Fuks Z et al (2000) Final
study of measurements and calculations of lung density report of the 70.2 and 75.6 Gy dose levels of a phase I dose
corrections for 6 and 18 MV photons. Int J Radiat Oncol escalation study using three-dimensional conformal radio-
Biol Phys 37:1163–1170 therapy in the treatment of inoperable non-small cell lung
Kong FM, Hayman JA, Griffith KA et al (2006) Final toxicity cancer. Cancer J 6:82–87
results of a radiation-dose escalation study in patients with Senan S, de Koste J, Samson M et al (1999) Evaluation of a
non-small-cell lung cancer (NSCLC): predictors for radia- target contouring protocol for 3D conformal radiotherapy in
tion pneumonitis and fibrosis. Int J Radiat Oncol Biol Phys non-small cell lung cancer. Radiother Oncol 53:247–255
65:1075–1086 Senan S, De Ruysscher D, Giraud P et al (2004) Literature-
Kubo HD, Len PM, Minohara S et al (2000) Breathing- based recommendations for treatment planning and execu-
synchronized radiotherapy program at the University of tion in high dose radiotherapy for lung cancer. Radiother
California Davis Cancer Center. Med Phys 27:346–353 Oncol 71:139–146
Kwa SL, Lebesque JV, Theuws JC et al (1998) Radiation Sepenwoolde Y, Lebesque JV, de Jaeger K et al (2003)
pneumonitis as a function of mean lung dose: an analysis of Comparing different NTCP models that predict the inci-
pooled data of 540 patients. Int J Radiat Oncol Biol Phys dence of radiation pneumonitis. Normal tissue complication
42:1–9 probability. Int J Radiat Oncol Biol 55:724–735
Mackie TR, el-Khatib E, Battista J et al (1985) Lung dose Sibley GS, Mundt AJ, Shapiro C et al (1995) The treatment of
corrections for 6 and 15 MV X rays. Med Phys 12:327–332 stage III nonsmall cell lung cancer using high dose
Marks LB, Munley MT, Bentel GC et al (1997) Physical and conformal radiotherapy. Int J Radiat Oncol Biol Phys
biological predictors of changes in whole-lung function 33:1001–1007
following thoracic irradiation. Int J Radiat Oncol Biol Phys Ten Haken RK, Martel MK, Kessler ML et al (1993) Use of
39:563–570 Veff and iso-NTCP in the implementation of dose escalation
Martel MK, Ten Haken RK, Hazuka MB et al (1994) Dose- protocols. Int J Radiat Oncol Biol Phys 27:689–695
volume histogram and 3-D treatment planning evaluation of Timmerman R, Papiez L, McGarry R et al (2003) Extracranial
patients with pneumonitis. Int J Radiat Oncol Biol Phys stereotactic radioablation: results of a phase I study in
28:575–581 medically inoperable stage I non-small cell lung cancer.
Martel MK, Ten Haken RK, Hazuka MB et al (1999) Chest 124:1946–1955
Estimation of tumor control probability model parameters Wang L, Yorke E, Chui CS (2002) Monte Carlo evaluation of
from 3-D dose distributions of non-small cell lung cancer 6 MV intensity modulated radiotherapy plans for head and
patients. Lung Cancer 24:31–37 neck and lung treatments. Med Phys 29:2705–2717
McShan DL, Fraass BA, Lichter AS (1990) Full integration of Wong JW, Sharpe MB, Jaffray DA et al (1999) The use of
the beam’s eye view concept into computerized treatment active breathing control (ABC) to reduce margin for
planning. Int J Radiat Oncol Biol Phys 18:1485–1494 breathing motion. Int J Radiat Oncol Biol Phys 44:911–919
Four-dimensional Radiation Therapy
for Non-Small Cell Lung Cancer:
A Clinical Perspective
Max Dahele, Johan Cuijpers, and Suresh Senan

Contents Abstract
The purpose of this chapter is to review 4-dimen-
1 Introduction.............................................................. 157 sional radiotherapy (4DRT) for lung cancer in a
2 What is Four-Dimensional Radiotherapy?........... 159 way that supports the wider adoption of existing
2.1 How Much Do Lung Tumors and Mediastinal technologies and effective treatment strategies.
Lymph Nodes Move? ................................................ 161 4DRT is defined, the components of a 4DRT
3 What Is the Evidence to Support the Use program are identified, resource-sensitive 4DRT
of 4DRT in Lung Cancer?...................................... 163 strategies are addressed and our own institution’s
4 An Institutional Approach to 4DRT ..................... 165 approach to 4D lung RT is discussed.
5 Can I Still Perform Image Guided 4DRT
without a 4DCT or Cone Beam CT? .................... 167
6 Strategies to Facilitate Knowledge Transfer 1 Introduction
and 4DRT Implementation..................................... 168
7 Conclusion ................................................................ 168 Translating existing knowledge and techniques into
References.......................................................................... 168 routine clinical use can present significant chal-
lenges, but is essential for improving the effective-
ness of radiotherapy (RT) and improving local
control and survival (Palma et al. 2010). Over the
last two decades a convergence of technologies has
helped to shape modern day four-dimensional
radiotherapy (4DRT) for lung cancer. Several of
these are highlighted in Table 1. However, at a time
when the prognosis for many patients with lung
cancer remains poor, many technical advances
have failed to enter routine clinical use, even
after the available technology has been acquired
(Mayles 2010; Routsis et al. 2010; Whitton et al.
2009). Although the explanations for this will likely
vary between healthcare systems, one possible rea-
son is that useful developments in RT are sometimes
M. Dahele (&)  J. Cuijpers  S. Senan seen as difficult to implement, or perceived as
Department of Radiation Oncology,
too complex or time consuming for day to day
VU University Medical Center, De Boelelaan 1117,
1081 HV, Amsterdam, The Netherlands use. Frequently, a lack of resources is implicated
e-mail: m.dahele@vumc.nl (Mayles 2010) however, all too often a systematic

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 157
DOI: 10.1007/174_2011_284, Ó Springer-Verlag Berlin Heidelberg 2011
158 M. Dahele et al.

Table 1 Over the last two decades a variety of technologies have contributed to modern day four-dimensional radiotherapy
(4DRT) several are highlighted here
Advances Comments
Three-dimensional conformal 3DCRT introduced about 20 years ago helped by computerized planning systems and multi-leaf
radiotherapy (3DCRT) collimators. Dose–volume histograms and multi-planar dose displays highlighted by Emami et al.
(1991). It was anticipated that ‘major progress towards uncomplicated local regional control of lung
cancer may be forthcoming’
Planning studies suggested 3DCRT could improve the therapeutic ratio for high-dose lung RT
(Armstrong et al. 1993)
Stereotactic lung RT Seminal report from Blomgren et al. (1995), over 15 years ago, described extracranial stereotactic RT
for targets in various locations including lung. With follow-up of 1.5–38 months local control was 80%
Intensity modulated RT (IMRT) Reports of IMRT for lung tumors appeared almost 15 years ago. In an early planning study, Derycke
et al. (1997) used non-coplanar intensity modulated beams to escalate the dose to central stage III
tumors
Concepts for current day tomotherapy and intensity modulated arc therapy techniques were described
in 1993 and 1995, respectively (Mackie et al. 1993; Yu 1995)
Moving beam treatments capable of rotation/arc delivery in any plane were being used over 50 years
ago to deliver doses comparable to the present time (Clarkson et al. 1959)
Respiratory gated RT (RGRT) Concept of gated radiation therapy demonstrated over 20 years ago (Ohara et al. 1989)
In mid-1999 Minohara et al. reported treatment of over 150 patients with lung or liver tumors using a
gated heavy-ion technique that incorporated an external infrared sensor to determine the respiratory
signal and digitized fluoroscopic images to assess organ motion (Minohara et al. 2000). The same
group reported the use of in-room CT for positional verification 12 years ago (Kamada et al. 1999)
Active breathing control (ABC) for temporarily suspending breathing and reducing tumor motion in
the thorax and abdomen described by Wong et al. (1999) and initial experience with deep inspiratory
breath hold RT for lung cancer by Rosenzweig et al. (2000), who also reported verbal commands could
increase the regularity of breathing and therefore improve the performance of respiratory gating
(Mageras et al. 2001)
Cone beam CT (CBCT) CBCT for radiotherapy described over 15 years ago (Cho et al. 1995). Jaffray et al. (2002) reported the
development of a high-resolution gantry mounted flat panel CBCT unit suitable for high-precision
image guided RT (IGRT)
Sonke et al. (2005) described 4D, respiratory correlated CBCT in 2005
Use of external markers to track lung Early studies seeking to characterize breathing induced tumor motion and its relationship to external
tumor motion markers were reported a decade ago and highlighted the challenges of phase discordance between
target and surrogate (Chen et al. 2001)
Computed tomography (CT) for Dosimetric problems associated with free breathing CT studies incorrectly localizing the position and
imaging tumor motion volume of critical structures described in the mid-1990s (Balter et al. 1996)
Six ‘fast’ CT scans may underestimate motion in early-stage tumors compared to 4DCT (Underberg
et al. 2004)
Slow CT described around 10 years ago. Without 4DCT tumor motion can be approximated using the
contour (plus a margin) from a single ‘slow’ CT scan limited to the region of interest (in combination
with a fast CT of the whole thorax for treatment planning) (Lagerwaard et al. 2001; Nakamura et al.
2008)
Slow CT is feasible with present-day scanners (Chinneck et al. 2010)
Two-phase shallow inspiration and expiration breath hold imaging can also be used when 4DCT is not
available (Ritchie et al. 1994; Yamada et al. 2002). This strategy was described over 15 years ago
4DCT has been in clinical use for close to a decade and workflows describing its use in patients with
early stage lung cancer have been published (Underberg et al. 2005). It has become the preferred CT
technique for routine imaging of mobile structures
Recent data from Riegel et al. (2009) suggest that cine CT may be an alternative to 4DCT—they
observed that it identified more motion than 4DCT in phantom studies
A new generation of 256- and 320-slice CT scanners is well suited to volumetric cine CT (Mori et al.
2007; Coolens et al. 2009)
(continued)
Four-dimensional Radiation Therapy 159

Table 1 (continued)
Advances Comments
Positron emission tomography 4D PET-CT may provide more complete information than 3D PET-CT about the motion of a
metabolically active region and result in less blurring (Aristophanous et al. 2011)
‘Optimal gating’ may quantify the maximum standardized uptake value (SUVmax) better than 3D PET
and produce images with less noise than 4D PET (van Elmpt et al. 2011)
Several studies have now evaluated the correlation between PET and pathology reaching various
conclusions about the optimum metric for target volume segmentation (Wanet et al. 2011; Devic et al.
2010) (note when evaluating such studies that the tumor is moving in vivo, but not ex vivo)
Heterogeneity corrected treatment Radiation treatment planning systems (RTP) with heterogeneity correction (HC) are now standard
planning systems (Elmpt et al. 2008). While they can better model the dose distribution in lung tumors and organs at risk
(OAR) no improvement in outcome has been directly attributed to their use
Caution with respect to tumor and OAR doses is needed when comparing non-HC and HC treatment
schedules or converting from non-HC to HC RTP systems (Franks et al. 2010; Hurkmans et al. 2009)
Multi-modal image fusion for In a 2009 survey of American radiation oncologists (36% responded), 95% reported using some form
treatment planning of advanced imaging in RTP for example PET (76%) and 4DCT (44%), most often for lung tumors
(Simpson et al. 2009)
Clear guidelines needed to support the optimum use of multi-modal imaging in RTP
Uncertainty margins Stereotactic lung RT typically uses reduced margins to minimize normal tissue toxicity. The high doses
immediately around the gross tumor volume (GTV) may permit this (Arvidson et al. 2008)
In the early phase of our own stereotactic lung practice local control was high with GTV–PTV margins
of only 3 mm, while at the same time setting up patients on the spine (Lagerwaard et al. 2008)
Clarity is needed before attributing improvements in outcome to technology rather than effective dose-
fractionation schedules in appropriately selected patients

and focused approach to change and implementation


may be lacking (Kotter 1995; Spear 2004). Under 2 What is Four-Dimensional
such conditions the risk is high that technology imple- Radiotherapy?
mentation projects will fail, deliver below expectation,
or take a very long time to complete. Ultimately this Four-dimensional lung RT makes it possible to
adversely affects their ability to impact positively on identify and account for breathing related tumor and
patient outcomes. (OAR) motion during radiation treatment plan-
Against this background, this chapter addresses the ning and delivery. This concept is not new as the
following practical issues: (1) key concepts in 4DRT, movement of lung tumors and surrounding struc-
(2) existing technical and educational resources, (3) tures was also appreciated during treatment simu-
components of a modern day lung RT program, (4) lation in the 2DRT era. Indeed many of the
the impact of 4DRT technologies, (5) our institution’s goals of 2D lung RT mirror those in 4DRT. For
approach to the routine clinical use of 4DRT, (6) example,
resources required for different 4DRT strategies, and 1. Identify target motion during imaging: 2DRT =
(7) strategies to promote knowledge transfer and the fluoroscopy at the simulator, 4DRT = 4D (retro-
implementation of 4DRT. Throughout this chapter we spective) respiratory- correlated computed tomog-
focus on what can be achieved in everyday clinical raphy (4DCT)
practice using standard commercial equipment. This 2. Account for motion during treatment planning
does not currently include true 4D dose computation, while trying to spare normal tissues: 2DRT =
which has lead some to suggest that full 4DRT has yet asymmetric margins to reflect typical tumor
to arrive. We have elected to focus on techniques and motion (largest in the cranio-caudal direction) and
technologies that reflect our institutional practice, but respect normal tissues, 4DRT = individualized
additional references describing other approaches to target volume incorporating respiratory motion,
4DRT are provided. No liability is assumed and one example of which is a motion encompass-
techniques are subject to change. ing internal target volume (ITV) with isotropic
160 M. Dahele et al.

Table 2 Selected resources that address important aspects of four-dimensional radiotherapy


Resources References (years)
EORTC recommendations for high-dose, high precision lung cancer RT De Ruysscher et al. (2010)
AAPM report on the management of respiratory motion Keall et al. (2006)
Tumor and normal tissue motion during breathing Weiss et al. (2007)
Review of 4D (computed tomography, CT) imaging and treatment planning Keall (2004)
Motion management of thoracic tumors during treatment planning and delivery Verellen et al. (2010)
Challenges in quality assurance for 4DRT Jiang et al. (2008)
4DCT quality assurance Hurkmans et al. (2011)
Clinical implementation of respiratory gated intensity modulated RT Keall et al. (2006)
Four-dimensional radiotherapy for lung cancer Lagerwaard and Senan (2007)
Review that links 4D and adaptive RT for lung cancer Sonke and Belderbos (2010)
Strategies to increase the conformity of lung RT, including 4DRT Chang and Cox (2010)
AAPM American Association of Physicists in Medicine, 4DRT Four-dimensional radiotherapy, 4DCT Four-dimensional computed
tomography, IMRT Intensity-modulated radiotherapy, 4DCBCT Four-cone beam computed tomography

expansion to create the planning target volume (e.g., CBCT) can also provide information about
(PTV) tumor motion, including a time-averaged volume that
3. Verify that initial RT fields are appropriate: approximates the tumor as seen on the average
2DRT = visual check prior to treatment using intensity projection (Ave-IP) of the 4DCT or full
fluoroscopy at the simulator, 4DRT = volumetric 4DCBCT, which allows for 4D image guided RT
on board imaging, for example 4D-cone beam CT (IGRT) delivery. CBCT can also provide information
(4DCBCT) or non-4DCBCT (may not completely about changes in tumor and OAR anatomy and
image motion) location that underpin adaptive radiotherapy (ART).
4. Verifying motion during a course of treatment: Again it is the name ART and the technology that is
2DRT = check fluoroscopy on the simulator, new, not the concept, which was also practiced
4DRT = range of approaches from no regular during the 2D era, for example a new immobilization
check, to some form of intermittent or daily online mask and treatment plan might be made when a head
imaging such as non-4D/4DCBCT or fluoroscopy. and neck tumor was clinically responding. A more
In 3D conformal RT (3DCRT), the use of con- detailed discussion of technical issues and the theory
ventional CT scans allowed for direct imaging of the of 4DRT and ART is found in recent publications
tumor and organs at risk, and led to improvements (Table 2).
over 2DRT in target coverage and OAR sparing From the preceding discussion, it can be appreci-
(Graham et al. 1994; Ragan and Perez 1978). How- ated that the availability of 4DCT, a radiation treat-
ever, conventional CT scans may contain substantial ment planning system (RTP)—preferably one with
motion artifact and they provide no information type B heterogeneity correction that adequately
about tumor displacement which risks both a geo- models photon and electron transport in low density
graphic target miss and the irradiation of excessive tissues (e.g., convolution/superposition, Monte Carlo,
amounts of normal tissue (Shih et al. 2004). The last or the Analytical Anisotropic Agorithm, ‘AAA’)
decade or so has resulted in the development of (Hurkmans et al. 2009), and a CBCT mounted on a
various commercial imaging solutions including standard linear accelerator fitted with a multi-leaf
4DCT that address this shortcoming (Slotman et al. collimator represents one common equipment con-
2006). These make it possible to routinely combine figuration that provides all the physical building
motion and detailed anatomical information in blocks for a fully-fledged 4D-adaptive lung RT pro-
order to create motion encompassing target volumes gram. Figure 1 puts 4D lung RT into the wider con-
(e.g., ITV) that can form the basis for treatment text of a comprehensive conventional and stereotactic
planning. Commercial onboard volumetric imaging lung RT program.
Four-dimensional Radiation Therapy 161

and in developing efficient workflows that allow for


the incorporation of essential 4D routines into daily
clinical practice. The practical question of whether a
single 4DCT is sufficient to identify tumor motion has
been addressed by several authors including Under-
berg et al. (2004) who compared a single ten-phase
4DCT with six fast multi-slice CT scans, Gucken-
berger et al. (2007) who evaluated repeated 4DCT
scans taken at 10 min-intervals and Redmond et al.
(2009) who compared initial 4DCT scans with two
subsequent rescans. The overall message from these
studies is that for most patients a single 4DCT is
probably acceptable although there are individual
patients with irregular breathing for whom this is not
the case (Fig. 2). 4DCT remains prone to artifact with
one retrospective study finding at least one artifact in
90% of patients (45/50) and 30% (6/20) of lung or
mediastinal tumors (Yamamoto et al. 2008). This
highlights that 4DCT scans must be carefully
reviewed at the time of acquisition (Fig. 3).
It is also clear that mediastinal lymph nodes move
(Fig. 4) although often to a different degree than the
primary tumor and not necessarily in the same phase.
Pantarotto et al. (2009) used 4DCT to study the motion
of 100 lymph nodes in 41 patients with either stages I or
III non-small cell lung cancer (NSCLC). They found
that 10% of nodes moved more than 1 cm, with those in
the lower mediastinum moving the most. Assessment
of the motion of 16 locally advanced tumors revealed
no association between primary tumor and nodal
motion, and phase offsets of at least 20 or 30% were
seen in 33 and 12% of nodes, respectively. In 11/16
patients at least one exhaustively trial primary tumor. In
Fig. 1 Elements of a contemporary lung cancer radiation
therapy program including Four-dimensional radiotherapy an ideal world, this argues for the inclusion of infor-
(4DRT) mation about both lymph nodes and the primary tumor
in adaptive radiotherapy strategies, especially when
2.1 How Much Do Lung Tumors using highly conformal plans. However, mediastinal
and Mediastinal Lymph Nodes structures are typically poorly visualized on cone beam
Move? computed tomography (CBCT). Donnelly et al. (2007)
have reported similar findings and Sher et al. (2007)
Four-dimensional radiotherapy for lung cancer is quantified mediastinal and hilar node motion using
predicated on the fact that lung tumors move. 4DCT in 24 patients and reported mean peak-to-peak
A number of studies have investigated the magnitude cranial-caudal motion in paratracheal, subcarinal, and
of this motion and two of these using 4DCT are hilar locations of 4, 6, and 7 mm, respectively . The
summarized in Table 3. In short, many tumors move routine use of 4DCT to contour nodal motion allows an
less than might be expected, usually less than 1 cm in ITV to be created for individual nodes during the
any direction. Clinical challenges include identifying planning process.
those tumors that move so much, or so erratically, that Once a treatment plan has been derived using
they risk compromising the effectiveness of treatment, patient-specific motion information, delivery may be
162 M. Dahele et al.

Table 3 Lung tumor motion evaluated with four-dimensional computed tomography (4DCT)
Author Institutions Comments
(Refs.)
Liu (2007) MD 166 tumors, 152 patients (57.2% stage III/IV) 4DCT normal respiration
Anderson 39.2, 1.8, 5.4% of tumors moved [0.5 cm along superior–inferior (SI), lateral and anterior–
posterior (AP) axes, respectively
95% of tumors moved \1.34, 0.4 and 0.59 cm along these axes
10.8% of tumors moved [1 cm along SI axis
Small tumors in lower half of lung more mobile
Motion driven mainly by movement of the diaphragm
Redmond Johns 20 patients
(2009) Hopkins Mean GTV excursion in SI, lateral and AP directions was 0.67, 0.21 and 0.29 cm, respectively

Fig. 2 Four-dimensional computed tomography for stereotac- blue 4DCT2 (generally smaller amplitude and in this case also
tic lung radiotherapy (RT). The upper and lower panels (left) less tumor motion), yellow fraction 1 (even smaller amplitude).
each show a blended image comprising the end-inspiration and This illustrates some of the challenges of 4DCT and shows that
end-expiration phases of separate 4DCT scans from the same for selected patients with variable breathing, one 4DCT may
patient taken at one imaging session. In the first (upper) study not be representative. In this situation relative uncertainty
of the whole thorax the cranio-caudal extent of tumor motion increases. In selected cases we have constructed an initial ITV
was approximately 2 cm. A second limited 4DCT of the tumor with information from both 4DCT studies, followed by online
region revealed cranio-caudal motion of 1.2 cm (lower). This monitoring of breathing using Real-time Position Management
variation is also reflected in different breathing patterns/ (RPM) system, (Varian Medical Systems Inc.) ± repeat 4DCT
amplitudes as shown by the traces on the right: pink 4DCT1, and re-planning as indicated

compromised in several ways, for example if there is co-location of the target and a critical structure may
an excessive change in the magnitude and direction of result in unintended dose being delivered to the OAR.
motion, or if the tumor position changes systemati- Although there are now several strategies for assess-
cally in relation to the planned location due to ing tumor motion and location before treatment
anatomical changes and baseline drift. An awareness delivery, it is not yet the standard practice to evaluate
of changes in tumor position relative to critical these parameters during beam-on, or to obtain a
structures is important when performing online dynamic online evaluation of the dosimetric conse-
target-based image-guidance, since a change in the quences of such changes.
Four-dimensional Radiation Therapy 163

Fig. 4 In this illustration, the mediastinum contains extensive


lymphadenopathy. On four-dimensional computed tomography
(4DCT) imaging the lateral border of the lymphadenopathy is
seen to move a maximum of approximately 1 cm in the medio-
lateral plane

tumor, in order to study any differences in patient


outcomes that might arise and determine efficacy and
cost-effectiveness. Specialists in other disciplines
might reasonably object to such a scenario. Further-
more, it is important that the resources required for a
clinical trial and the altruism of patients are used to
address the most appropriate clinical questions in
Fig. 3 The upper image illustrates multi-level 4DCT artifact such a way that an answer is likely to be forthcoming,
(arrows). When there is artifact in the region of the tumor or an and to consider that clinical trials may not always be
adjacent critical structure we typically acquire a second short
4DCT that can be used for contouring these volumes (lower the most appropriate way of using scarce resources
image shows superimposed full thorax and limited volume that in this case might perhaps be better deployed to
scans). Although at times a 4DCT study of the full thorax may implement effective treatments like stereotactic body
need to be repeated, the initial average intensity projection radiotherapy (SBRT) for lung cancer (Palma et al.
(Ave-IP) is frequently acceptable to be used for dose calcula-
tion and contouring the remaining organs at risk 2010; De Ruysscher et al. 2010) or to make full use of
expensive equipment that has already been purchased.
There is a risk that studies designed to look at the
benefit of a specific technology will be expensive,
3 What Is the Evidence to Support underpowered, deliver out of date results, and deflect
the Use of 4DRT in Lung Cancer? attention and resources away from day-to-day priori-
ties and other major barriers to implementing effec-
The pragmatic view is that one of the main aims of tive treatments in under-resourced environments
RT is to hit the target whilst avoiding as much normal (Nagata et al. 2011). The idea that not to exhaustively
tissue damage as possible in the process. Nonetheless, trial technologies means a market free for all does not
even when a technology provides better information stand up to scrutiny. As we have illustrated, basic
about the target (e.g., 4DRT, IGRT) there have been equipment that has been commercially available for
calls for high-level evidence to support its use. On the best part of 5–10 years is what is required to do
one level this seems similar to asking a surgeon to 4D-adaptive lung RT, and if acquiring such equip-
operate with inferior equipment that compromises ment is not possible, less sophisticated variants of
their chance of performing a complete excision of the 4DRT may still be possible.
164 M. Dahele et al.

Table 4 Outcomes for locally advanced non-small cell lung cancer (NSCLC) treated with today’s advanced technologies are
comparable to those reported in the preceding era, as are the delivered doses
Study (Refs.) Years Radiation arm of interest Outcomes
RTOG 9410 (three-arm phase III 1994–1998 63 Gy/34fr (1.8 Gy 9 25, Median survival (MS) 17 m with
study) (Curran et al. 2003) then 2 Gy 9 9) minimum and median ‘potential’
follow-up (FU) times of 4 and 6 years
Concurrent chemo-RT 21% 4 year survival
Stages II and III Total of 595 evaluable patients in
Two-phase (boost) three arms
technique
CT-based treatment
planning whenever possible
SWOG 9504 (single arm phase II 1996–1998 61 Gy/33fr (1.8 Gy 9 25, MS of 26 m for 83 eligible patients
study) (Gandara et al. 2003) then 2 Gy 9 8) with median FU 32 m
Two phase RT using post- 1, 2, and 3 year survival rates 76, 54
induction volume for phase and 37%
two)
Concurrent chemo-RT
Proven stage IIIB
2 dimensional planning
Sura et al. (2008) (retrospective 2001–2005 Stages I–IIIB inoperable MS 25 m for stage III, with median
institutional review) Patients CT-planned and FU 26 m among survivors
treated with IMRT using
1.8–2 Gy/fr
Minimum dose 60 Gy, 2 year overall survival 58%
mean 69.5 Gy for stage III
23/39 stage III pts had
sequential chemo-RT and
13/39 concurrent chemo-
RT
Liao et al. (2010) (retrospective 1999–2006, Concurrent chemo-RT Mean FU 3DCRT = 2.1 years, mean
institutional review comparing 4DCT ? IMRT FU 4DCT/IMRT = 1.3 years
CT ? 3DCRT with introduced 2004 3DCRT = 318pts, 4DCT/ Median survival 0.85 years (10.2 m)
4DCT ? IMRT) IMRT = 91pts for 3DCRT and 1.4 years (16.8 m)
for 4DCT/IMRT
Both groups median dose Reduced grade C3 radiation
63 Gy pneumonitis with 4DCT/IMRT
Percentage of N2/3 in
3DCRT group 60/27% and
4DCT/IMRT group 48/32%
Percentage with PET
staging 3DCRT 49 versus
82% in 4DCT/IMRT

The growing availability of advanced technologies trend to reduced target volumes brought about by a
including 4DRT (Table 1) raises the issue of whether shift away from elective nodal irradiation (Table 4)
it is having a discernible effect in the clinic. If we look (De Ruysscher et al. 2010; Dillman et al. 1990, 1996;
at locally advanced lung cancer one conclusion might Kong et al. 2005). One study in Table 4 deserves
be that although techniques have changed this is not more attention (Liao et al. 2010). Liao et al. (2010)
reflected in the prescribed dose or clinical outcomes, have suggested that for patients treated with chemo-
despite evidence of a dose-response relationship and a radiation, a more recent cohort whose radiotherapy
Four-dimensional Radiation Therapy 165

incorporated a combination of 4DCT and IMRT did the Varian ‘AAA’ algorithm for dose calculation
better than a group treated with 3DCRT both in terms (EclipseTM TPS, Varian Medical Systems Inc.). For
of overall survival and reduced lung toxicity. Whilst conventional chemo-radiotherapy, a dose of 66 Gy in
these intriguing data allow for hypothesis generation, 33 fractions of 2 Gy is delivered in eligible patients
when they are interpreted in the context of other with locally advanced tumors (Lagerwaard et al.
available studies they perhaps raise at least as many 2008; Phernambucq et al. 2011).
questions as they answer (Gandara et al. 2003; Inoue For 4DCT imaging, patients enter the scanner and
et al. 2001; Bentzen 2008; Yom et al. 2007; Albain are settled by the technologist who explains the pro-
et al. 2002; Phernambucq et al. 2011). Indeed, it cedure. Both groups of patients lie on a thin mattress
remains to be seen whether the inference that placed on top of the treatment couch, with a foam
advanced technologies per se caused the gain in sur- cushion under the knees and an adjustable support to
vival is supported, or whether they were one factor comfortably place the arms above the head. If this is
associated with it. not possible, an alternative position has to be used,
So where does this leave things? Our own depart- such as one arm up, and one arm down, or both arms
ment, which operates within the constraints of a down. Patients to be treated with stereotactic lung RT
public healthcare system, aims to create an environ- have infrared surface markers placed on their thorax
ment that is receptive to the rapid adoption of new (ExacTracÒ, Brainlab AG, Feldkirchen, Germany).
technologies and techniques that can make a specific Once the patient is breathing comfortably, a ten-phase
treatment possible (Lagerwaard et al. 2008), permit free-breathing 4DCT is acquired using a 16-slice
the delivery of potentially toxic treatment with a wide-bore CT scanner (GE DiscoveryTM CT590 RT,
lower predicted risk (Verbakel et al. 2010), or enable GE Healthcare, Waukesha, WI, USA) and the Varian
greater efficiency (Verbakel et al. 2009). Technology Real-time Position ManagementTM (RPM) system
is an enabler but the most important component of the (Varian Medical Systems Inc.) (Underberg et al.
treatment chain remains the radiation and how it is 2004). After 4DCT acquisition the online recon-
used. For example, during the first 5 years of our structed 4D scan is reviewed by the technologists for
department’s stereotactic lung RT program, early- artifacts and the patient’s respiratory trace is analyzed
stage peripheral lung tumors were treated using for reproducibility. During this time the patient
4DCT-based imaging in combination with patient remains in the treatment position on the couch. If an
setup on the spine (Lagerwaard et al. 2008). This imaging artifact is observed in the tumor region, or
produced results comparable to that reported by the CT has been acquired at an unrepresentative point
groups using 4DCT with online CBCT for localiza- in the breathing trace, then the scan is repeated, either
tion based on the tumor position (Taremi et al. 2011). a complete scan of the thorax or a shorter one that
We currently use non-4DCBCT-based online setup, includes the target lesion with a cranio-caudal margin
and have changed our delivery technique from 8–12 of several centimeters (Fig. 3) (Guckenberger et al.
non co-planar fixed beams to volumetric modulated 2007; Yamamoto et al. 2008). In the latter scenario,
arc therapy (RapidArcÒ, Varian Medical Systems the long scan that encompasses the whole thorax will
Inc., Palo Alto, CA, USA). be used for dose calculation and lung volumes and the
shorter scan for target delineation (± adjacent critical
structures). Skin marks are made to guide initial
4 An Institutional Approach to 4DRT patient positioning prior to treatment delivery.
For 4D lung SBRT, ITV is defined using the maxi-
4DCT imaging was introduced into our department in mum intensity projection (MIP), modified as necessary
2003, and all patients with early stage and locally using information from the individual phases (modified
advanced tumors are planned using this technique. MIP ITV) (Underberg et al. 2005; Lagerwaard
There are alternative strategies, but Fig. 5 illustrates et al. 2008). If the MIP is considered unreliable, for
the key steps that are currently used for both groups of example, when the tumor is in close proximity to the
patients. The present dose/fractionation schedules are chest wall or other solid structures, then individual
based upon a ‘risk-adapted’ philosophy for stereo- phases are relied upon. Mediastinal and lung window
tactic RT: 3 9 18, 5 9 11 and 8 9 7.5 Gy using settings are used to aid the contouring process. Either
166 M. Dahele et al.

Fig. 5 Flowchart illustrating


the approach to four-
dimensional radiotherapy
(4DRT) for stereotactic
(left hand images) and
conventional (right hand
images) lung radiotherapy in
our institution. Row 1 =
four-dimensional computed
tomography (4DCT) imaging,
Row 2 = target volume
delineation, Row 3 =
low- dose region (5 Gy), Row
4 = high-dose region (95%),
Row 5 = online cone beam
CT (CBCT) target and planar
kilovoltage (kV) spine
imaging. Individual steps
are described further and
referenced in the text

all ten phases can be used or only those phases that contours are attached to the Ave-IP dataset that is used
represent the extremes of motion in each translational for dose calculation and normal structure delineation
direction can be selected by viewing the moving tumor (Admiraal et al. 2008). Where necessary certain mobile
on an appropriate 4D workstation. If contouring is organs are also contoured using 4DCT (e.g., the
performed using a system other than the RTP, then it esophagus, or for some lower zone tumors the stomach
should be verified as being correct after transfer to the or bowel) (Dieleman et al. 2007). Automated contour
RTP, as changes in the position of the original contours propagation is not yet in routine use (van Dam et al.
of up to several millimeters have occasionally been 2010) and experience so far suggests such tools require
identified. For routine cases a 5 mm isotropic ITV– careful visual inspection ± manual adjustment.
PTV margin is currently used. A dosimetric margin is in Motion suppression, gating, tracking, and implan-
the process of being introduced for tumors with a ted fiducials are not currently used for stereotactic
motion amplitude [15 mm (Cuijpers et al. 2010). All lung RT at our center. Patients are treated under free
Four-dimensional Radiation Therapy 167

breathing conditions. Initial positioning is using orthogonal X-ray imaging and intermittent CBCT.
skin marks with ExacTracÒ markers ± stereoscopic At the minimum, CBCT imaging is acquired at the
X-rays with 6D robotic table corrections (e.g. for beginning of weeks 1, 3, and 5 (of a 33 fraction
tumors close to the spine). A non-gated CBCT schedule, treating with five daily fractions per week)
(On-Board ImagerÒ, Varian Medical Systems Inc.) is however, this may be increased depending for
acquired and an automatic target match is performed example on the presence of atelectasis or other fea-
using the Ave-IP planning CT as the reference data tures. It is important to note that certain changes in
set, followed by visual verification of target alignment normal anatomy and tumor position can be detected
in all three planes and manual adjustment as neces- using planar imaging provided that the team is aware
sary. For selected tumors very close to the spine the of what to look for (e.g., tracheal shift, aided by the
patient may be set up to the spine, to ensure that the trachea contour, or the development of consolidation).
spinal cord dose is as planned. The position of Daily IGRT can help to control systematic and ran-
the target and critical structures is verified using the dom errors and allow a reduction in setup margins
ITV/PTV and critical structure contours, and where (Higgins et al. 2010; Yeung et al. 2010). After treat-
necessary the relevant critical structure isodose (e.g., ment the CBCT scans are visible in the treatment
the dose limit for the spinal cord). Infrared ExacTracÒ planning system (EclipseTM, Varian Medical Systems
markers are used for intra-fraction motion monitoring. Inc.) where they have been automatically registered
All patients are now treated with RapidArcÒ volu- with the planning CT. This means that as long as that
metric modulated RT, routinely consisting of two arcs was the position in which the patient was treated in
(Verbakel et al. 2009). Initial work has not demon- and provided that the anatomy is stable and no sig-
strated a clinically significant interplay effect between nificant new clinical changes have developed, it is
tissue motion and dynamic MLC movements (Ong possible to estimate dose coverage of the target and
et al. 2011). Individual patient dosimetric quality selected other structures. Certain isodoses (e.g., 95%)
assurance is performed using a commercial digital are also exported to the treatment unit for this purpose
2D ionization chamber array (MatriXX Evolu- (their use is subject to the same conditions). When
tion ± Gantry Angle Sensor, IBA Dosimetry GmbH, target volume coverage appears to be unsatisfactory,
Schwarzenbruck, Germany). Prior to being approved the radiation oncologist and physicist review the
by the radiation oncologist, all treatment plans are treatment plan and dosimetric coverage before
checked by one technologist and one physicist, who deciding on whether to acquire a new 4DCT and
are independent of the team that created the plan. replan the patient. In some cases a plan is calculated
In locally advanced non-small cell lung cancer, an on the CBCT as part of the decision- making process.
ITV consisting of the primary tumor and lymph nodes
are contoured using the extremes of motion identified
as above. The MIP data set is not used because the 5 Can I Still Perform Image Guided
ability to discriminate the boundary between tumor 4DRT without a 4DCT or Cone
and normal structures is typically lost in the medias- Beam CT?
tinal and hilar regions. Once again the Ave-IP is used
as the primary data set and contouring is performed In appropriately selected patients, alternatives to
on the phase bins representing the extreme positions 4DCT for identifying tumor motion include limited
of the tumor and lymph nodes, using mediastinal and volume slow CT, breath-hold imaging at normal end
lung window settings. An isotropic 10 mm ITV–PTV expiratory/inspiratory positions and repeated fast CT
expansion is typically used and most patients are (Underberg et al. 2004; Lagerwaard et al. 2001;
treated under free breathing conditions. If gating is Nakamura et al. 2008; Chinneck et al. 2010; Ritchie
used it is typically phase gating at end-inspiration et al. 1994; Yamada et al. 2002). An additional fast
with audio coaching. An increasing number of CT of the full thorax is also acquired with either of
plans are made using a hybrid conventional-IMRT the first two strategies to be used for treatment plan-
technique that has enabled a reduction in lung ning. Whilst the slow CT strategy can be used for
doses (Verbakel et al. 2010). Daily online setup stereotactic lung RT it is not suited to locally
is performed on the spine using expiration-gated advanced tumors, however, expiratory/inspiratory
168 M. Dahele et al.

breath-hold imaging remains an option. A motion national efforts such as those being rolled out in Canada
encompassing ITV can be created from the CT and the UK with the aim of increasing the uptake of
information with a further expansion to generate the specific technologies, in this case IMRT. Such pro-
PTV. If there is no access to CT then conventional grams typically have several components including the
simulator techniques can also be used to estimate development of standards, summaries of the available
visible tumor motion. In suitable patients with evidence, mentorship schemes, and targets (Whitton
peripheral lesions stereotactic lung treatment can be et al. 2009; Williams et al. 2010). How effective various
delivered under free-breathing conditions using pla- strategies turn out to be will be of great interest.
nar image-guidance [e.g., orthogonal kilovoltage (kV)
or megavoltage (MV) images] with patient setup on
the spine. Note that prior to commencing CBCT-
7 Conclusion
based tumor matching in 2008 for stereotactic RT of
peripheral lung tumors, we used stereoscopic kilo-
4DRT is an improvement on conventional 3DRT per-
voltage imaging to set up the patient on the spine and
formed without patient-specific motion information.
achieved high rates of local control (Lagerwaard et al.
The essential components of 4D lung RT can be
2008), which have stood the test of longer follow up.
implemented into routine clinical workflows and per-
Furthermore, the ITV was defined using an uncoached
formed using standard linear accelerators with a mini-
free breathing 4DCT, and a 3 mm ITV–PTV expan-
mum of additional equipment. Although the
sion was used, patients were treated in free breathing
contribution that 4DRT per se is making to clinical
with multiple non-coplanar beams, calculated using
outcomes is unresolved we feel that this is not a reason
the pencil beam algorithm with simple heterogeneity
to delay the implementation of logical and welcome
corrections (Lagerwaard et al. 2008). For patients
advances in radiotherapy, especially if they can assist
with locally advanced lung cancer and an appropriate
the development and delivery of more effective treat-
PTV, daily planar imaging with setup on the spine
ment schedules to more patients. Responsible and
appears to be acceptable, again treating without
effective purchasing of equipment and strategies to
respiratory coaching or gating (Table 4, Phernambucq
ensure that new equipment is brought quickly into
et al. 2011).
routine clinical use and used to facilitate the delivery of
the most effective treatments to as many patients as
possible are an important part of ensuring the maximum
6 Strategies to Facilitate Knowledge impact on patient care, return on investment, and the
Transfer and 4DRT Implementation efficient use of available resources.
Technology adoption and healthcare innovation pre- Conflicts of interest The Department of Radiation Oncology,
sents real challenges (Herzlinger 2006). It involves VU University Medical Center has research collaborations with
individual factors as diverse as beliefs, ambition, and Varian Medical Systems Inc. (Palo Alto, CA, USA), Brainlab
AG (Feldkirchen, Germany), Velocity Medical Solutions
ability to teamwork, and institutional factors such as
(Atlanta, GA, USA).
leadership, vision, focus, and people—who you hire,
how they are deployed, and how hard they work for the
group. At a departmental level, it requires clarity of
thought and purpose to identify and prioritize key
References
technologies and techniques that will result in the
ability to deliver the best treatments to patients, and Admiraal MA, Schuring D, Hurkmans CW (2008) Dose
calculations accounting for breathing motion in stereotactic
strategies to acquire essential knowledge and expertise, lung radiotherapy based on 4D-CT and the internal target
demystify outwardly complex techniques, and gain the volume. Radiother Oncol 86(1):55–60
confidence to use them (Dahele and Slotman 2011). Albain KS, Crowley JJ, Turrisi AT III, Gandara DR, Farrar
When it comes to implementation and change there are WB, Clark JI, Beasley KR, Livingston RB (2002) Concur-
rent cisplatin, etoposide, and chest radiotherapy in patho-
helpful resources that can provide practical guidance logic stage IIIB non-small-cell lung cancer: a Southwest
and motivation (Kotter 1995; Spear 2004; Sirkin et al. Oncology Group phase II study, SWOG 9019. J Clin Oncol
2005). This may be complemented by regional and 20(16):3454–3460
Four-dimensional Radiation Therapy 169

Aristophanous M, Berbeco RI, Killoran JH, Yap JT, Sher DJ, planning and delivery of high-dose, high-precision radio-
Allen AM, Larson E, Chen AB (2011) Clinical Utility of 4D therapy for lung cancer. J Clin Oncol 28(36):5301–5310
FDG-PET/CT Scans in Radiation Treatment Planning. Int J Derycke S, Van Duyse B, De Gersem W, De Wagter C, De
Radiat Oncol Biol Phys [Epub ahead of print]http://dx. Neve W (1997) Non-coplanar beam intensity modulation
doi.org/10.1016/j.ijrobp.2010.12.060 allows large dose escalation in stage III lung cancer.
Armstrong JG, Burman C, Leibel S, Fontenla D, Kutcher G, Radiother Oncol 45(3):253–261
Zelefsky M, Fuks Z (1993) Three-dimensional conformal Devic S, Tomic N, Faria S, Menard S, Lisbona R, Lehnert S
radiation therapy may improve the therapeutic ratio of high- (2010) Defining radiotherapy target volumes using 18F-
dose radiation therapy for lung cancer. Int J Radiat Oncol fluoro-deoxy-glucose positron emission tomography/com-
Biol Phys 26(4):685–689 puted tomography: still a Pandora’s box? Int J Radiat Oncol
Arvidson NB, Mehta MP, Tomé WA (2008) Dose coverage Biol Phys 78(5):1555–1562
beyond the gross tumor volume for various stereotactic Dieleman EM, Senan S, Vincent A, Lagerwaard FJ, Slotman
body radiotherapy planning techniques reporting similar BJ, de Koste JR (2007) Four-dimensional computed
control rates for stage I non-small-cell lung cancer. Int J tomographic analysis of esophageal mobility during
Radiat Oncol Biol Phys 72(5):1597–1603 normal respiration. Int J Radiat Oncol Biol Phys 67(3):
Balter JM, Ten Haken RK, Lawrence TS, Lam KL, Robertson 775–780
JM (1996) Uncertainties in CT-based radiation therapy Dillman RO, Seagren SL, Propert KJ, Guerra J, Eaton WL,
treatment planning associated with patient breathing. Int J Perry MC, Carey RW, Frei EF III, Green MR (1990) A
Radiat Oncol Biol Phys 36(1):167–174 randomized trial of induction chemotherapy plus high-dose
Bentzen SM (2008) Radiation oncology health technology radiation versus radiation alone in stage III non-small-cell
assessment: the best is the enemy of the good. Nat Clin lung cancer. N Engl J Med 323(14):940–945
Pract Oncol 5(10):563 Dillman RO, Herndon J, Seagren SL, Eaton WL Jr, Green MR
Blomgren H, Lax I, Näslund I, Svanström R (1995) Stereotactic (1996) Improved survival in stage III non-small-cell
high-dose fraction radiation therapy of extracranial tumors lung cancer: seven-year follow-up of cancer and leukemia
using an accelerator. Clinical experience of the first thirty- group B (CALGB) 8433 trial. J Natl Cancer Inst 88(17):
one patients. Acta Oncol 34(6):861–870 1210–1215
Chang JY, Cox JD (2010) Improving radiation conformality in Donnelly ED, Parikh PJ, Lu W, Zhao T, Lechleiter K, Nystrom
the treatment of non-small cell lung cancer. Semin Radiat M, Hubenschmidt JP, Low DA, Bradley JD (2007) Assess-
Oncol 20(3):171–177 ment of intrafraction mediastinal and hilar lymph node
Chen QS, Weinhous MS, Deibel FC, Ciezki JP, Macklis RM movement and comparison to lung tumor motion using
(2001) Fluoroscopic study of tumor motion due to breath- four-dimensional CT. Int J Radiat Oncol Biol Phys 69(2):
ing: facilitating precise radiation therapy for lung cancer 580–588
patients. Med Phys 28(9):1850–1856 Elmpt W, Ollers M, Velders M, Poels K, Mijnheer B,
Chinneck CD, McJury M, Hounsell AR (2010) The potential Ruysscher DD, Dekker A, Lambin P, Boersma L (2008)
for undertaking slow CT using a modern CT scanner. Br J Transition from a simple to a more advanced dose
Radiol 83(992):687–693 calculation algorithm for radiotherapy of non-small cell
Cho PS, Johnson RH, Griffin TW (1995) Cone-beam CT for lung cancer (NSCLC): implications for clinical implemen-
radiotherapy applications. Phys Med Biol 40(11):1863– tation in an individualized dose–escalation protocol. Radio-
1883 ther Oncol 88(3):326–334
Clarkson JR, Leech HJ, Taylor AG, Mason SW (1959) A Emami B, Purdy JA, Manolis J, Barest G, Cheng E, Coia L,
moving-beam caesium 137 telecurie unit. Br J Radiol Doppke K, Galvin J, LoSasso T, Matthews J et al (1991)
32:798–804 Three-dimensional treatment planning for lung cancer. Int J
Coolens C, Breen S, Purdie TG, Owrangi A, Publicover J, Radiat Oncol Biol Phys 21(1):217–227
Bartolac S, Jaffray DA (2009) Implementation and charac- Franks KN, Purdie TG, Dawson LA, Bezjak A, Jaffray DA,
terization of a 320-slice volumetric CT scanner for simu- Bissonnette JP (2010) Incorporating heterogeneity correc-
lation in radiation oncology. Med Phys 36(11):5120–5127 tion and 4DCT in lung stereotactic body radiation therapy
Cuijpers JP, Verbakel WF, Slotman BJ, Senan S (2010) A (SBRT): The effect on target coverage, organ-at-risk doses,
novel simple approach for incorporation of respiratory and dose conformity. Med Dosim 35(2):101–107
motion in stereotactic treatments of lung tumors. Radiother Gandara DR, Chansky K, Albain KS, Leigh BR, Gaspar LE,
Oncol 97(3):443–448 Lara PN Jr, Burris H, Gumerlock P, Kuebler JP, Bearden JD
Curran WJ, Scott CB, Langer CJ (2003) Long-term benefit is III, Crowley J, Livingston R (2003) Southwest Oncology
observed in a phase III comparison of sequential vs Group. Consolidation docetaxel after concurrent chemora-
concurrent chemoradiation for patients with unresected diotherapy in stage IIIB non-small-cell lung cancer: phase II
stage III NSCLC: RTOG 9410 (Abstract). Proc Am Soc Southwest Oncology Group Study S9504. J Clin Oncol
Clin Oncol 22:621 21(10):2004–2010
Dahele M, Slotman B (2011) Implementing new radiotherapy Graham MV, Matthews JW, Harms WB Sr, Emami B, Glazer
techniques. Br J Healthc Manag Guide Healthc Technol HS, Purdy JA (1994) Three-dimensional radiation treatment
17(3):22–30 planning study for patients with carcinoma of the lung. Int J
De Ruysscher D, Faivre-Finn C, Nestle U, Hurkmans CW, Le Radiat Oncol Biol Phys 29(5):1105–1117
Péchoux C, Price A, Senan S (2010) European Organisation Guckenberger M, Wilbert J, Meyer J, Baier K, Richter A,
for Research and Treatment of Cancer recommendations for Flentje M (2007) Is a single respiratory correlated 4D-CT
170 M. Dahele et al.

study sufficient for evaluation of breathing motion? Int J Lagerwaard FJ, van Sornsen De Koste JR, Nijssen-Visser MR,
Radiat Oncol Biol Phys 67(5):1352–1359 Schuchhard-Schipper RH, Oei SS, Munne A, Senan S
Herzlinger RE (2006) Why innovation in health care is so hard. (2001) Multiple ‘‘slow’’ CT scans for incorporating lung
Harv Bus Rev 84(5):58–66 156 tumor mobility in radiotherapy planning. Int J Radiat Oncol
Higgins J, Bezjak A, Hope A, Panzarella T, Li W, Cho JB, Biol Phys 51(4):932–937
Craig T, Brade A, Sun A, Bissonnette JP (2010) Effect of Lagerwaard FJ, Haasbeek CJ, Smit EF, Slotman BJ, Senan S
Image-Guidance Frequency on Geometric Accuracy and (2008) Outcomes of risk-adapted fractionated stereotactic
Setup Margins in Radiotherapy for Locally Advanced Lung radiotherapy for stage I non-small-cell lung cancer. Int J
Cancer. Int J Radiat Oncol Biol Phys [Epub ahead of print] Radiat Oncol Biol Phys 70(3):685–692
http://dx.doi.org/10.1016/j.ijrobp.2010.04.006 Liao ZX, Komaki RR, Thames HD Jr, Liu HH, Tucker SL,
Hurkmans CW, Cuijpers JP, Lagerwaard FJ, Widder J, van der Mohan R, Martel MK, Wei X, Yang K, Kim ES,
Heide UA, Schuring D, Senan S (2009) Recommendations Blumenschein G, Hong WK, Cox JD (2010) Influence of
for implementing stereotactic radiotherapy in peripheral technologic advances on outcomes in patients with unre-
stage IA non-small cell lung cancer: report from the Quality sectable, locally advanced non-small-cell lung cancer
Assurance Working Party of the randomised phase III receiving concomitant chemoradiotherapy. Int J Radiat
ROSEL study. Radiat Oncol 4:1 Oncol Biol Phys 76(3):775–781
Hurkmans CW, van Lieshout M, Schuring D, van Heumen MJ, Liu HH, Balter P, Tutt T, Choi B, Zhang J, Wang C, Chi M,
Cuijpers JP, Lagerwaard FJ, Widder J, van der Heide UA, Luo D, Pan T, Hunjan S, Starkschall G, Rosen I, Prado K,
Senan S (2011) Quality assurance of 4D-CT scan techniques Liao Z, Chang J, Komaki R, Cox JD, Mohan R, Dong L
in multicenter phase iii trial of surgery versus stereotactic (2007) Assessing respiration-induced tumor motion and
radiotherapy (Radiosurgery or surgery for operable early internal target volume using four-dimensional computed
stage (Stage 1A) non-small-cell Lung cancer [ROSEL] tomography for radiotherapy of lung cancer. Int J Radiat
Study). Int J Radiat Oncol Biol Phys 80(3):918–927 Oncol Biol Phys 68(2):531–540
Inoue A, Kunitoh H, Sekine I, Sumi M, Tokuuye K, Saijo N Mackie TR, Holmes T, Swerdloff S, Reckwerdt P, Deasy JO,
(2001) Radiation pneumonitis in lung cancer patients: a Yang J, Paliwal B, Kinsella T (1993) Tomotherapy: a new
retrospective study of risk factors and the long-term concept for the delivery of dynamic conformal radiotherapy.
prognosis. Int J Radiat Oncol Biol Phys 49(3):649–655 Med Phys 20(6):1709–1719
Jaffray DA, Siewerdsen JH, Wong JW, Martinez AA (2002) Mageras GS, Yorke E, Rosenzweig K, Braban L, Keatley E,
Flat-panel cone-beam computed tomography for image- Ford E, Leibel SA, Ling CC (2001) Fluoroscopic evaluation
guided radiation therapy. Int J Radiat Oncol Biol Phys of diaphragmatic motion reduction with a respiratory gated
53(5):1337–1349 radiotherapy system. J Appl Clin Med Phys 2(4):191–200
Jiang SB, Wolfgang J, Mageras GS (2008) Quality assurance Mayles WP (2010) Radiotherapy Development Board Survey of
challenges for motion-adaptive radiation therapy: gating, the availability and use of advanced radiotherapy technology
breath holding, and four-dimensional computed tomogra- in the UK. Clin Oncol (R Coll Radiol) 22(8):636–642
phy. Int J Radiat Oncol Biol Phys 71(1 suppl):S103–S107 Minohara S, Kanai T, Endo M, Noda K, Kanazawa M (2000)
Kamada T, Tsujii H, Mizoe JE, Matsuoka Y, Tsuji H, Osaka Y, Respiratory gated irradiation system for heavy-ion radio-
Minohara S, Miyahara N, Endo M, Kanai T (1999) A therapy. Int J Radiat Oncol Biol Phys 47(4):1097–1103
horizontal CT system dedicated to heavy-ion beam treat- Mori S, Endo M, Komatsu S, Yashiro T, Kandatsu S, Baba M
ment. Radiother Oncol 50(2):235–237 (2007) Four-dimensional measurement of lung tumor dis-
Keall P (2004) 4 dimensional computed tomography imaging placement using 256-multi-slice CT-scanner. Lung Cancer
and treatment planning. Semin Radiat Oncol 14(1):81–90 56(1):59–67
Keall PJ, Mageras GS, Balter JM, Emery RS, Forster KM, Jiang Nagata Y, Wulf J, Lax I, Timmerman R, Zimmermann F,
SB, Kapatoes JM, Low DA, Murphy MJ, Murray BR, Ramsey Stojkovski I, Jeremic B (2011) Stereotactic radiotherapy of
CR, Van Herk MB, Vedam SS, Wong JW, Yorke E (2006a) primary lung cancer and other targets: results of consultant
The management of respiratory motion in radiation oncology meeting of the International Atomic Energy Agency. Int J
report of AAPM Task Group 76. Med Phys 33(10):3874–3900 Radiat Oncol Biol Phys 79(3):660–669
Keall P, Vedam S, George R, Bartee C, Siebers J, Lerma F, Nakamura M, Narita Y, Matsuo Y, Narabayashi M, Nakata M,
Weiss E, Chung T (2006b) The clinical implementation of Yano S, Miyabe Y, Matsugi K, Sawada A, Norihisa Y,
respiratory-gated intensity-modulated radiotherapy. Med Mizowaki T, Nagata Y, Hiraoka M (2008) Geometrical
Dosim 31(2):152–162 differences in target volumes between slow CT and 4D CT
Kong FM, Ten Haken RK, Schipper MJ, Sullivan MA, Chen M, imaging in stereotactic body radiotherapy for lung tumors in
Lopez C, Kalemkerian GP, Hayman JA (2005) High-dose the upper and middle lobe. Med Phys 35(9):4142–4148
radiation improved local tumor control and overall survival Ohara K, Okumura T, Akisada M, Inada T, Mori T, Yokota H,
in patients with inoperable/unresectable non-small-cell lung Calaguas MJ (1989) Irradiation synchronized with respira-
cancer: long-term results of a radiation dose escalation tion gate. Int J Radiat Oncol Biol Phys 17(4):853–857
study. Int J Radiat Oncol Biol Phys 63(2):324–333 Ong C, Verbakel WF, Cuijpers JP, Slotman BJ, Senan S (2011)
Kotter JP (1995) Leading change: why transformation efforts Dosimetric impact of interplay effect on RapidArc lung
fail. Harv Bus Rev 73(2):59–67 stereotactic treatment delivery. Int J Radiat Oncol Biol Phys
Lagerwaard FJ, Senan S (2007) Lung cancer: intensity-mod- 79(1):305–311
ulated radiation therapy, four-dimensional imaging and Palma D, Visser O, Lagerwaard FJ, Belderbos J, Slotman BJ,
mobility management. Front Radiat Ther Oncol 40:239–252 Senan S (2010) Impact of introducing stereotactic lung
Four-dimensional Radiation Therapy 171

radiotherapy for elderly patients with stage I non-small-cell Spear SJ (2004) Learning to lead at Toyota. Harv Bus Rev
lung cancer: a population-based time-trend analysis. J Clin 82(5):78–86 151
Oncol 28(35):5153–5159 Sura S, Gupta V, Yorke E, Jackson A, Amols H, Rosenzweig
Pantarotto JR, Piet AH, Vincent A, de Koste JR, Senan S KE (2008) Intensity-modulated radiation therapy (IMRT)
(2009) Motion analysis of 100 mediastinal lymph nodes: for inoperable non-small cell lung cancer: the Memorial
potential pitfalls in treatment planning and adaptive strat- Sloan-Kettering Cancer Center (MSKCC) experience.
egies. Int J Radiat Oncol Biol Phys 74(4):1092–1099 Radiother Oncol 87(1):17–23
Phernambucq EC, Spoelstra FO, Verbakel WF, Postmus PE, Taremi M, Hope A, Dahele M, Pearson S, Fung S, Purdie T,
Melissant CF, Maassen van den Brink KI, Frings V, van de Brade A, Cho J, Sun A, Bissonnette JP, Bezjak A (2011)
Ven PM, Smit EF, Senan S (2011) Outcomes of concurrent Stereotactic body radiotherapy for medically inoperable
chemoradiotherapy in patients with stage III non-small-cell lung cancer: prospective, single-center study of 108 con-
lung cancer and significant comorbidity. Ann Oncol secutive patients. Int J Radiat Oncol Biol Phys [Epub ahead
22(1):132–138 of print]
Ragan DP, Perez CA (1978) Efficacy of CT-assisted two Underberg RW, Lagerwaard FJ, Cuijpers JP, Slotman BJ, de
dimensional treatment planning: analysis of 45 patients. Koste JR, Senan S (2004) Four-dimensional CT scans for
AJR Am J Roentgenol 131(1):75–79 treatment planning in stereotactic radiotherapy for stage I
Redmond KJ, Song DY, Fox JL, Zhou J, Rosenzweig CN, Ford lung cancer. Int J Radiat Oncol Biol Phys 60(4):1283–1290
E (2009) Respiratory motion changes of lung tumors over Underberg RW, Lagerwaard FJ, Slotman BJ, Cuijpers JP,
the course of radiation therapy based on respiration- Senan S (2005) Use of maximum intensity projections
correlated four-dimensional computed tomography scans. (MIP) for target volume generation in 4DCT scans for lung
Int J Radiat Oncol Biol Phys 75(5):1605–1612 cancer. Int J Radiat Oncol Biol Phys 63(1):253–260
Riegel AC, Chang JY, Vedam SS, Johnson V, Chi PC, Pan T van Dam IE, de Koste JR, Hanna GG, Muirhead R, Slotman BJ,
(2009) Cine computed tomography without respiratory Senan S (2010) Improving target delineation on 4 dimen-
surrogate in planning stereotactic radiotherapy for non- sional CT scans in stage I NSCLC using a deformable
small-cell lung cancer. Int J Radiat Oncol Biol Phys registration tool. Radiother Oncol 96(1):67–72
73(2):433–441 van Elmpt W, Hamill J, Jones J, De Ruysscher D, Lambin P,
Ritchie CJ, Hsieh J, Gard MF, Godwin JD, Kim Y, Crawford CR Ollers M (2011) Optimal gating compared to 3D and 4D
(1994) Predictive respiratory gating: a new method to reduce PET reconstruction for characterization of lung tumours.
motion artifacts on CT scans. Radiology 190(3):847–852 Eur J Nucl Med Mol Imaging 38(5):843–855
Rosenzweig KE, Hanley J, Mah D, Mageras G, Hunt M, Toner Verbakel WF, Senan S, Cuijpers JP, Slotman BJ, Lagerwaard
S, Burman C, Ling CC, Mychalczak B, Fuks Z, Leibel SA FJ (2009) Rapid delivery of stereotactic radiotherapy for
(2000) The deep inspiration breath-hold technique in the peripheral lung tumors using volumetric intensity-modu-
treatment of inoperable non-small-cell lung cancer. Int J lated arcs. Radiother Oncol 93(1):122–124
Radiat Oncol Biol Phys 48(1):81–87 Verbakel W, Ladenius-Lischer I, Slotman BJ, Senan S (2010)
Routsis D, Staffurth J, Beardmore C, Mackay R (2010) A hybrid IMRT technique for high-dose radiotherapy in
Radiotherapy Development Board Education and training large-volume stage III lung tumors: planning comparison
for intensity-modulated radiotherapy in the UK. Clin Oncol with 3 other techniques. Int J Radiat Oncol Biol Phys
(R Coll Radiol) 22(8):675–680 78(3):S811 (A3357)
Sher DJ, Wolfgang JA, Niemierko A, Choi NC (2007) Verellen D, Depuydt T, Gevaert T, Linthout N, Tournel K,
Quantification of mediastinal and hilar lymph node move- Duchateau M, Reynders T, Storme G, De Ridder M (2010)
ment using four-dimensional computed tomography scan: Gating and tracking, 4D in thoracic tumours. Cancer
implications for radiation treatment planning. Int J Radiat Radiother 14(6-7):446–454
Oncol Biol Phys 69(5):1402–1408 Wanet M, Lee JA, Weynand B, De Bast M, Poncelet A, Lacroix
Shih HA, Jiang SB, Aljarrah KM, Doppke KP, Choi NC (2004) V, Coche E, Grégoire V, Geets X (2011) Gradient-based
Internal target volume determined with expansion margins delineation of the primary GTV on FDG-PET in non-small
beyond composite gross tumor volume in three-dimensional cell lung cancer: a comparison with threshold-based
conformal radiotherapy for lung cancer. Int J Radiat Oncol approaches, CT and surgical specimens. Radiother Oncol
Biol Phys 60(2):613–622 98(1):117–125
Simpson DR, Lawson JD, Nath SK, Rose BS, Mundt AJ, Mell Weiss E, Wijesooriya K, Dill SV, Keall PJ (2007) Tumor and
LK (2009) Utilization of advanced imaging technologies for normal tissue motion in the thorax during respiration:
target delineation in radiation oncology. J Am Coll Radiol analysis of volumetric and positional variations using 4D
6(12):876–883 CT. Int J Radiat Oncol Biol Phys 67(1):296–307
Sirkin HL, Keenan P, Jackson A (2005) The hard side of Whitton A, Warde P, Sharpe M, Oliver TK, Bak K, Leszczyn-
change management. Harv Bus Rev 83(10):108–118 158 ski K, Etheridge S, Fleming K, Gutierrez E, Favell L, Green
Slotman BJ, Lagerwaard FJ, Senan S (2006) 4D imaging for E (2009) Organisational standards for the delivery of
target definition in stereotactic radiotherapy for lung cancer. intensity-modulated radiation therapy in Ontario. Clin
Acta Oncol 45(7):966–972 Oncol (R Coll Radiol) 21(3):192–203
Sonke JJ, Belderbos J (2010) Adaptive radiotherapy for lung Williams MV, Cooper T, Mackay R, Staffurth J, Routsis D,
cancer. Semin Radiat Oncol 20(2):94–106 Burnet N (2010) The implementation of intensity-modu-
Sonke JJ, Zijp L, Remeijer P, van Herk M (2005) Respiratory lated radiotherapy in the UK. Clin Oncol (R Coll Radiol)
correlated cone beam CT. Med Phys 32(4):1176–1186 22(8):623–628
172 M. Dahele et al.

Wong JW, Sharpe MB, Jaffray DA, Kini VR, Robertson JM, Yeung AR, Li J, Shi W, Newlin H, Morris CG, Samant S, Saito
Stromberg JS, Martinez AA (1999) The use of active A, Chvetsov A, Liu C, Palta J, Olivier K (2010) Optimal
breathing control (ABC) to reduce margin for breathing image-guidance scenario with cone-beam computed tomog-
motion. Int J Radiat Oncol Biol Phys 44(4):911–919 raphy in conventionally fractionated radiotherapy for lung
Yamada K, Soejima T, Yoden E, Maruta T, Okayama T, tumors. Am J Clin Oncol 33(3):276–280
Sugimura K (2002) Improvement of three-dimensional Yom SS, Liao Z, Liu HH, Tucker SL, Hu CS, Wei X, Wang X,
treatment planning models of small lung targets using Wang S, Mohan R, Cox JD, Komaki R (2007) Initial
high-speed multi-slice computed tomographic imaging. Int J evaluation of treatment-related pneumonitis in advanced-
Radiat Oncol Biol Phys 54(4):1210–1216 stage non-small-cell lung cancer patients treated with
Yamamoto T, Langner U, Loo BW Jr, Shen J, Keall PJ concurrent chemotherapy and intensity-modulated radio-
(2008) Retrospective analysis of artifacts in four-dimen- therapy. Int J Radiat Oncol Biol Phys 68(1):94–102
sional CT images of 50 abdominal and thoracic radio- Yu CX (1995) Intensity-modulated arc therapy with dynamic
therapy patients. Int J Radiat Oncol Biol Phys 72(4): multileaf collimation: an alternative to tomotherapy. Phys
1250–1258 Med Biol 40(9):1435–1449
PET and PET/CT in Treatment Planning
Michael P. Mac Manus and Rodney J. Hicks

Contents Abstract
Positron emission tomography (PET) is a major
1 Introduction.............................................................. 173 advance in lung cancer imaging and is having an
2 Lessons for the Radiation Oncologist From increasing impact on the management of patients
Preoperative PET Staging in Potentially with non-small cell lung cancer who are candidates
Resectable NSCLC .................................................. 174 for potentially-curative treatment with radiother-
2.1 Intra-Thoracic Lymph Node Evaluation................... 174
apy. PET imaging, using 18F-flurodeoxyglucose as
2.2 Evaluation for Distant Metastasis ............................. 174
the tracer, and more recently in the form of FDG-
3 Role of PET in Selecting Patients for PET/CT is now the most important single imaging
Radiotherapy/Chemoradiotherapy in NSCLC..... 175
modality for staging, patient selection and radio-
4 Use of PET and PET/CT for Radiotherapy therapy planning in NSCLC. If scans are acquired
Treatment Planning................................................. 177
under appropriate conditions and the patient is
5 Tumor Contouring .................................................. 178 positioned for radiotherapy, a single scan can be
6 Role PET in Restaging After Definitive Radical used for all of these purposes. In this chapter the
Radiotherapy/Chemoradiotherapy for NSCLC ... 179 role of PET and PET/CT in staging, patient
7 The Future of PET in NSCLC .............................. 181 selection and radiotherapy planning are discussed.
7.1 Use of PET Tracers Other Than FDG in Staging Additionally, the use of FDG-PET for response
and Treatment Response Assessment in Lung assessment is described and finally the potential
Cancer ........................................................................ 181
value of PET tracers other than FDG is considered.
7.2 Respiratory Gating of PET Data............................... 183
7.3 Response Adapted Radiotherapy: Targeting Tumor
Subvolumes During a Course of Radiotherapy........ 183
8 Conclusions ............................................................... 184
1 Introduction
References.......................................................................... 184

The advent of positron emission tomography (PET)


has been the most significant advance in cancer
M. P. Mac Manus (&)
Department of Radiation Oncology, imaging since the introduction of computed tomog-
Peter MacCallum Cancer Centre, raphy (CT) (Stroobants et al 2003) and is having an
St Andrew’s Place, East Melbourne, increasing impact on the management of patients with
VIC 3002, Australia lung cancer, especially those being considered for
e-mail: Michael.macmanus@petermac.org
radiotherapy (Hicks and Mac Manus 2003). The
R. J. Hicks problems caused by separate acquisition of PET and
Centre for Molecular Imaging,
Peter MacCallum Cancer Centre, CT images are becoming increasingly rare as hybrid
St Andrew’s Place, East Melbourne, PET/CT scanners supersede older stand-alone PET
VIC 3002, Australia

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 173
DOI: 10.1007/174_2011_300, Ó Springer-Verlag Berlin Heidelberg 2011
174 M. P. Mac Manus and R. J. Hicks

scanners (Townsend and Beyer 2002). PET/CT has sensitivity and specificity for the detection of lymph
become an essential investigation for the radiation node involvement by tumor (Toloza et al. 2003).
oncologist, both helping to exclude from radical Numerous clinicopathological studies and meta-
radiotherapy those patients who cannot benefit because analyses have proven the superior accuracy of PET
they have extensive thoracic or distant metastatic dis- staging for mediastinal involvement compared to CT
ease and serving as the primary tool for target volume based staging. In the Gould meta-analysis of 39
definition, based on a growing literature of prospective studies with surgical confirmation of imaging results,
studies and expert reviews. PET/CT can minimize median sensitivity and specificity for PET were 85
the risk of a geographic miss while on average reducing and 90%, respectively (Gould et al. 2003). By con-
the unnecessary irradiation of normal tissues and trast, sensitivity of 57% and specificity of 82% were
demonstrating a higher degree of reproducibility reported for CT (Toloza et al. 2003). Mediastinal CT
compared to CT-based planning. Since the last edition is falsely negative in the approximately 20% of
of this book, the evidence base for the use of PET-based patients with non-enlarged lymph nodes but approx-
staging and radiotherapy planning has further imately 80% of patients with pathologically positive
strengthened and, despite the absence of randomized normal-sized nodes had positive PET findings in those
trials in radiotherapy patients, PET/CT is widely con- nodes. This information is very useful for planning
sidered to be a mandatory investigation for patients radiotherapy with PET/CT. Nevertheless, both tech-
with non-small cell lung cancer (NSCLC) who are niques can miss small volume nodal deposits that can
candidates for potentially curative radiotherapy. only be identified by pathological examination. Sur-
The literature supporting use of FDG PET and vival in NSCLC is powerfully correlated with lymph
PET/CT in small cell lung cancer (SCLC) is still sparse, node staging and drops precipitously when mediasti-
although PET can frequently influence management nal nodes contain tumor. Dunagan and colleagues
decisions (Bradley et al. 2004; Blum et al. 2004). reported that survival was more strongly correlated
Accordingly, most of the literature on the value of PET with PET stage than CT based stage in a large group
scanning in lung cancer relates to NSCLC. The role of of patients, who were mostly surgical candidates
PET and PET/CT in evaluation of patients who are (Dunagan et al. 2001). We have found similarly
planned to receive radical radiotherapy will be dis- superior prognostic stratification by FDG PET in a
cussed in this chapter. A single PET/CT scan can per- group of patients with more advanced conventional
form dual roles for the radiation oncologist; (1) staging stage, many of which were only candidates for
for patient selection for radiotherapy and (2) determi- radiotherapy (Hicks et al. 2001).
nation of the gross tumor volume (GTV) for treatment
planning. These roles are inseparable because the
staging information used in patient selection is also 2.2 Evaluation for Distant Metastasis
employed for target volume definition and therefore
both will be considered together in this chapter. PET can typically detect unsuspected distant metastasis
in 5–10% of patients with potentially-resectable stages
I–II disease but the rate of detection is higher in those
2 Lessons for the Radiation with more loco-regionally advanced disease. Both PET
Oncologist From Preoperative PET and PET/CT are capable of accurately detecting disease
Staging in Potentially Resectable in the adrenals. PET is also more specific than radio-
NSCLC nuclide bone scanning in the detection of bone
metastasis in lung cancer. In a study from Peter
2.1 Intra-Thoracic Lymph Node MacCallum Cancer Institute of 42 patients with
Evaluation PET-detected distant metastasis before planned surgery
(n = 7) or radical radiotherapy (RT)/chemoradiother-
When CT is used to stage the mediastinum, node apy (n = 35) for NSCLC, survival was investigated as
size is the only significant parameter to be consid- the principal endpoint (MacManus et al. 2003). The
ered, commonly using a short axis diameter of 1 cm influence of metastasis number and other prognostic
to indicate malignancy. Accordingly CT has poor factors was investigated using Cox regression analysis.
PET and PET/CT in Treatment Planning 175

All but four patients had died by last follow up. Prognostic stratification is an important, albeit
Median survival was 9 months overall, 12 months for indirect, means of assessing the relative diagnostic
27 patients with single PET-detected metastasis performance of different staging procedures. At Peter
and 5 months for 15 patients with [1 metastasis MacCallum Cancer Centre, a prospective study was
(p = 0.009). ECOG performance status (p = 0.027) but instituted in 1996 in which 153 consecutive patients
not pre-PET stage, weight loss or metastasis site corre- with NSCLC who were candidates for radical radio-
lated with survival. Dual modality PET/CT staging is therapy, in most cases given with concurrent che-
more accurate for detection of distant metastasis in motherapy, underwent both conventional staging and
NSCLC than either PET or CT as single modalities. FDG-PET prior to therapy (Mac Manus et al. 2001).
Accurate localization of FDG avid regions on fused CT After PET, 30% of patients were denied radical
and PET images reduces the risk of false positives. radiotherapy because of unexpected distant metastasis
(Fig. 1) or because of PET-detected intrathoracic
disease that was too extensive for safe radical irradi-
3 Role of PET in Selecting Patients ation. In the 107 patients who actually underwent
for Radiotherapy/ radical therapies, PET stage powerfully correlated
Chemoradiotherapy in NSCLC with survival (p = 0.0041) whereas conventional
stage correlated rather poorly with survival
Radical radiotherapy or chemoradiotherapy is offered (p = 0.19). Early in this study, PET findings of
predominantly to suitable patients who have stage IIIA extensive disease that were unsupported by review of
or IIIB disease. Patients with stages I–II disease who conventional imaging were not judged to be sufficient
cannot undergo resection because of significant co- reason to deny a patient an attempt at radical therapy.
morbidity may also be treated with radical radiother- However, it soon became clear that early progression
apy. The factors that make these patients unsuitable for occurred at all untreated metastatic sites detected by
surgery (advanced disease or significant comorbidi- PET. In separate study from Peter MacCallum, it was
ties), militate against confirmation of their intratho- reported that conventional T and N stage assessment
racic lymph node status at thoracotomy. These factors in patients treated with radical radiotherapy is a
consequently necessitate accurate non-invasive meth- relatively poor predictor of outcome (Ball et al. 2002),
ods of staging. For pragmatic reasons, the literature which is in contrast to the situation surgically treated
evaluating the diagnostic performance of FDG PET patients who are better served by the current staging
has primarily focused on staging prior to surgery in system. Our experience using PET/CT for staging
cohorts of patients with predominantly stages I–II radiotherapy patients over the past decade suggests
disease since histopathologic staging provides an that approximately a third of patients being consid-
accepted reference standard for staging accuracy. As ered for radical treatment are unsuitable for this
detailed above, such studies have consistently indi- treatment. Similar experience has been recently
cated the superiority of PET and PET/CT compared to reported by a Polish group who found that only 75 of
CT. However, there is no reason to suppose that FDG 100 patients with NSCLC being considered for radi-
PET or PET/CT are any less reliable in stage III dis- cal RT actually went on to receive this treatment after
ease, despite the relative paucity of studies in which PET/CT imaging (Kolodziejczyk et al. 2011). In
rigorous pathological confirmation has been per- particular, detection of distant metastatic disease is
formed in this setting. Because it is generally neither critical to avoiding futile attempts at curative radio-
ethical or feasible to subject such patients to compre- therapy, which involves considerable expense,
hensive biopsy of all PET and CT detected lesions resource utilization and toxicity. As expected on
simply to assess diagnostic accuracy, the only practical Bayesian principles, the likelihood of metastatic
means to compare diagnostic performance is to per- disease increases with increasing conventional stage.
form detailed follow-up using serial imaging and sur- In a cohort of 167 patients, the rate of PET-detected
vival as reference standards. Based on these standards, metastasis increased significantly (p = 0.016) with
FDG-PET is clearly the most reliable non-invasive increasing pre-PET stage from I (7.5%) through II
staging test, and justifies its use in the staging of (18%) to III (24%), and, in particular, was signifi-
patients who are candidates for radical radiotherapy. cantly higher in stage III (p = 0.039) than I–II.
176 M. P. Mac Manus and R. J. Hicks

Fig. 1 Following diagnostic


CT and evaluation of
pulmonary function tests this
patient was considered to be a
radical radiotherapy candidate
and therefore underwent FDG
PET/CT scanning on a flat
palette in treatment position.
Although mediastinal nodes
were mildly enlarged, only
similar size nodes in the
superior right hilum had
significant FDG-avidity.
However, intense focal
abnormality in the L-3
vertebra (fused PET/CT in
right upper panel and PET
in left lower panel) without
corresponding CT
abnormality (left middle
panel) indicated metastatic
bone disease. There were also
several other metabolically-
active bone marrow
abnormalities including
lesions in the left posterior
iliac crest, right lesser
trochanter and right ischium
(arrowed on the maximum
intensity pixel (MIP)
projection image in the lower
left panel) and a volume
rendered image (right upper
panel). Accordingly, the
patient was considered to
have stage IV disease and the
treatment plan was changed
to palliative radiotherapy

Eschmann and colleagues reported a similarly high The impact of PET selection on survival of
rate of detection of distant metastases in apparent patients treated with radical radiotherapy has been
stage III disease (Eschmann et al. 2007). These data illustrated by a further study from Peter MacCallum
have significant implications for radiotherapy patients Cancer Centre in which two prospective cohorts with
in whom conventional stage is generally more and without access to PET staging were compared
advanced than in surgical cohorts and also limits the (Mac Manus et al. 2002). Cohort 1 consisted of all
ability to directly extrapolate data of diagnostic participants in an Australian randomized trial from
accuracy of FDG PET and PET/CT from surgical our center given 60 Gy conventionally fractionated
series to impute potential costs and benefits in radical radiotherapy with or without concurrent car-
radiotherapy settings although this is often attempted boplatin from 1989 to 1995. Eligible patients had
(Buck et al. 2010). Stages I–III, Eastern Cooperative Oncology Group
PET and PET/CT in Treatment Planning 177

status 0 or 1, \10% weight loss, and had not under- normal tissues. Although this ideal is usually unat-
gone PET. Cohort 2 included all radical radiotherapy tainable, because the RT dose needed to control the
candidates between November 1996 and April 1999 majority of lung cancers is greater than the dose that
who received RRT after PET staging and fulfilled the can be given while maintaining the function of sur-
same criteria for stage, Eastern Cooperative Oncology rounding normal tissues, complementary advances in
Group status, and weight loss. Eighty and seventy molecular imaging and radiotherapy delivery mean
seven eligible patients comprised the PET and that significant improvements in locoregional disease
non-PET groups, respectively. The median survival control are possible. PET/CT based planning is an
was 31 months for PET patients and 16 months for exciting development that will help maximize the
non-PET patients. Mortality from NSCLC in the first benefit from advanced radiotherapy techniques, which
year was 17% for PET patients and 32% for non-PET are often associated with tight margins around tumors,
patients, respectively. The hazard ratio for NSCLC by ensuring that the treatment accurately targets all
mortality for PET versus non-PET patients was 0.49 sites of gross disease.
(p = 0.0016) on unifactorial analysis and was 0.55 The burgeoning PET and PET/CT literature clearly
(p = 0.0075) after adjusting for chemotherapy, which shows that an estimate of disease extent based on
significantly improved survival. This study suggests FDG-PET is very frequently different from and is
that, by using PET to exclude unsuitable patients with usually more accurate than an estimate made using
advanced disease and by integrating it within the CT alone. Because pathological confirmation of the
radiotherapy treatment planning process, previously truth of either PET/CT or CT based staging is rarely
unattainable survival results can be obtained. available in stage III NSCLC patients treated with
As the central importance of PET in selecting radical radiotherapy, it is reasonable to employ PET/
patients for radical radiotherapy for NSCLC becomes CT information as a de facto ‘‘gold standard’’ for
established, it is becomes important to consider the radiotherapy planning. In a growing number of
timeliness of the scans used for radiotherapy plan- studies in the literature, PET or PET/CT based
ning. In some countries with restricted access to radiotherapy planning has been compared with CT
radiotherapy services, a significant period may elapse alone plans, using each patient as their own control.
between FDG-PET scanning and initiation of RT. In a Comparisons between RT treatment plans or tumor
prospective study at our centre, we reported a very volumes made with and without the assistance of PET
high rate of disease progression between initial stag- have been made.
ing PET scans and RT planning images performed a PET based radiotherapy planning has been shown to
median of 3 weeks later. Approximately 39% of scans have an especially high impact on patients with
showed progressive disease at loco-regional or distant atelectasis. On CT scans, atelectatic lung and tumor
metastatic sites, which in 29% of cases caused a tissues typically have similar densities. The lack of
change to from radical palliative therapy (Everitt et al. contrast makes it impossible for the radiation oncolo-
2010). Thus, if there is any significant delay between gist to do anything other than guess where the bound-
FDG PET/CT and delivery of radiotherapy, it is likely ary between tumor and lung lies. However, when CT
that progression of disease may invalidate either the information is supplemented by FDG PET informa-
treatment plan or intent. tion, especially in the form of a fused PET/CT image, it
is often very easy to determine the boundary between
normal tissue and tumor, thereby attaining tumor
4 Use of PET and PET/CT coverage with the least exposure of uninvolved lung.
for Radiotherapy Treatment Prospective and retrospective trials in which PET
Planning and PET/CT have been used for RT planning in lung
cancer are listed in Table 1. All reports have shown a
The intention behind radical or ‘‘definitive’’ radio- clinically significant impact of PET, although most
therapy in NSCLC is cure but with acceptable toxic- studies were small. The effect of PET on radiotherapy
ity. An ideal radiotherapy plan would deliver planning varied between studies, at least in part
sufficient radiation to control the tumor whilst because of variations in sample size and treatment
depositing less than tolerance dose to surrounding practices at the various institutions. In cases with
178 M. P. Mac Manus and R. J. Hicks

Table 1 Studies of PET or PET/CT in RT planning with [20 patients


Author/year Patients Parameter studied Change in RT parameter
Munley et al. (1999) 35 RT field [34%
Nestle et al. (1999) 34 RT field 35%
Vanuytsel et al. (2000) 73 GTV 62%
Caldwell et al. (2001) 30 GTV 100%
Mac Manus et al. (2001) 102 GTV 38%
Mah et al. (2002) 23 PTV 100%
Bradley et al. (2004) 24 GTV 58%
Van Der Wel et al. (2005) 21 GTV 100%
RT field 100%
De Ruysscher et al. (2005) 21 RT dose deliverable with equal toxicity 55.2 Gy RT
68.9 Gy PET
Deniaud-Alexandre et al. (2005) 92 GTV 48%
Van Loon et al. (2008) 21 Change in RT plan (SCLC) 24%
Pommier et al. (2010) 119 Change in RT plan 31%
Kolodziejczyk et al. (2011) 75 Geographic miss 27% if PET not used
Abbreviations: RT Radiotherapy, GTV gross tumour volume, PTV planning target volume, SCLC small cell lung cancer

atelactasis (Nestle et al. 1999) the target volume was


often reduced because non-cancerous atelectatic lung 5 Tumor Contouring
was excluded from the treatment volume. In cases
with more extensive nodal involvement than sus- The topic of tumor contouring (definition of the
pected on CT, PET could lead to an increase in target margin of the tumor in 3 dimensional (3D) space) in
volume (Mac Manus et al. 2001). At some centers, NSCLC is a complex one. Early studies of the use of
including our own, enlarged but FDG negative nodes CT images to define target volumes for RT in lung
are routinely excluded from the GTV, unless there are cancer were characterized by poor agreement between
clear features on CT that the nodes are largely observers when they were asked to contour the same
necrotic. Other centers regard such nodes as suspi- tumors (van de Steene et al. 2002). There was both
cious based only on size criteria and irradiate them. very wide inter- and intra- observer variability
Different planning policies can therefore have a large because the CT scans contained insufficient data to
influence on the impact of PET. allow a clear delineation of tumor extent. However, it
Despite differences in methodology, all published is possible to improve upon this by ensuring that
studies of PET and PET/CT in radiotherapy planning observers adhere to a defined contouring protocol
show that this new technology has a very significant (Bowden et al. 2002). Fused PET and CT images are
impact on treatment volumes. It is currently impos- much more suitable for contouring tumors because
sible to quantify the benefit of PET based planning, they provide greater contrast between tumor and
but it is reasonable to assume that a treatment plan normal tissues. Nevertheless, even with PET/CT
made using a more accurate estimate of tumor extent images significant variation between observers can
is going to be a better one. One significant conse- occur if steps are not taken to ensure that a stan-
quence of the use of PET and PET/CT for radio- dardized contouring process is used. One cause of
therapy planning is the need for the PET suite to be difficulty is the relative low resolution of PET com-
considered as part of the chain of radiotherapy quality pared to CT, leading to blurring of the margins of
control. Technical aspects of the use of PET and PET/ structures apparent on PET scans. At our own centre,
CT in radiotherapy planning were considered in detail we adapted our established CT contouring protocol to
in the IAEA expert report (MacManus et al. 2009). apply it to PET/CT based planning and found that a
PET and PET/CT in Treatment Planning 179

detailed visual contouring protocol could achieve a space within which it moves. Effectively a 3D space
high level of reproducibility between observers that contains all of the tumor, all or most of the time is
(Bayne et al. 2010). This process gave the observers produced. Margins are added for microscopic disease
consistent instructions for window settings and image (clinical target volume or CTV) and this is used to
display and provided rules for contouring nodes and define a larger planning target volume (PTV) which is
determining tumor margins. It relied upon the spatial actually treated. The volume enclosed by a moving
abilities of the human brain to synthesize information tumor with an additional margin has been referred to
from the PET and CT components of the images and as an ‘‘internal target volume’’ or ITV. The prolonged
all other clinical information. acquisition time of PET provides a more accurate
Other groups have used automated contouring estimate of the average location of the tumor over
software approaches that rely upon mathematical time than a single CT image.
functions to objectively determine the tumor margins.
These methods use only the FDG PET data and do not
take into account the CT information but have the 6 Role PET in Restaging After
advantage that they should give identical results each Definitive Radical Radiotherapy/
time they are used on a particular imaging dataset. Chemoradiotherapy for NSCLC
The wide range of automated methods available is an
indication that the problems raised by this approach Response to therapy could potentially determine the
have not been solved. The same tumor (Nestle et al. further management of patients with lung cancer after
2005) may appear to vary greatly in size depending radical radiotherapy, including consideration for sal-
upon which method is used there is no consistently vage or consolidation therapies in incomplete or
effective method that can reliably account for tumor complete responders. Three-dimensional structural
movement, even in phantoms (Yaremko et al. 2005) imaging modalities, such as CT and MRI have long
All of the automated methods require editing by a been the most important investigations for assessment
human observer, but because of their reproducibility of response to non-surgical therapies such as radiation
they may provide an excellent starting point for sub- therapy or chemotherapy. World Health Organization
sequent editing and thereby reduce variation between or RECIST criteria are applied to measurements of
observers. Some centers have used simple SUV cut- tumor dimensions made before and after therapy and
offs but these are especially unreliable (Biehl et al. responses are categorized as complete response (CR),
2006) and will give very different results depending partial response (PR), no response (NR) or progres-
upon the level of SUV selected (Hong et al. 2007). sive disease but are recognized to have significant
More complex methods such as tumor to background limitations (Jaffe 2006). In NSCLC in particular,
ratio analysis are to be preferred (Nestle et al. 2006). tumors may be obscured by atelectasis before or after
Nevertheless the location of a moving tumor in 3D therapy and may be obscured by radiation pneumo-
space cannot be verified independently so no reliable nitis or fibrosis in the post treatment period (Lever
gold standard exists. Histopathological analysis of et al. 1984). As discussed above, lymph node size
resected lung specimens (van Baardwijk et al. 2007) measured on CT is an unreliable measure of lymph
and subsequent comparison with preoperative imag- node involvement by tumor. Tumors often regress
ing are plagued by inevitable distortions and con- gradually over several months, mandating serial
tractions that occur when tissues are processed for measurements to assess response. Lesions may
pathological examination (Dahele et al. 2008). Anal- never regress radiologically despite having been
ysis of patterns of failure may ultimately provide the controlled by treatment. FDG-PET may facilitate
only clinically relevant measure of the accuracy of more accurate early assessment of response to treat-
PET based radiotherapy planning. ment of NSCLC than structural imaging. Preliminary
The primary step in radiotherapy planning is to evidence in patients that PET scanning may be useful
contour all gross tumor deposits. Because of the after radiation therapy and it is probably superior to
effects of movement with respiration the process of CT for detecting both the presence and the extent of
GTV definition aims to describe not just the location recurrent disease irrespective of the primary treatment
of tumor at a particular instant, but to delineate the modality.
180 M. P. Mac Manus and R. J. Hicks

Prospective data from Peter MacCallum Cancer Although there is scientific appeal in the absolute
Centre (Mac Manus et al. 2003), show that a visually evaluation of glucose metabolic rate in tumors, fully
read PET response is much more powerfully corre- quantitative approaches using arterial blood analysis
lated with survival than response measured by CT are probably too invasive and complex for routine
scanning. Seventy-three patients with NSCLC clinical use. In clinical trials of novel therapeutic
underwent PET and CT scans before and after radical agents, semi-quantitative measures based on the
radiotherapy (n = 10) or chemoradiotherapy change in standardized uptake (SUV) in lesions can
(n = 63). Follow-up PET scans were performed at a be reasonably well standardized if attention is paid to
median of 70 days post radiotherapy. Each patient study acquisition parameters (Binns et al. 2011).
had prospective determinations of response to therapy Whichever approach has been chosen, a reduction in
made with PET and CT. In this study a visual FDG uptake generally predicts patient benefit in
assessment was made to determine PET-response patients treated for lung cancer (Hicks 2009). Chan-
while WHO criteria were used for CT response. PET- ges in SUV have been shown to have prognostic
response categories were defined as follows; significance after neoadjuvant chemotherapy prior to
(1) CMR (complete metabolic response)—no abnor- surgery and after palliative chemotherapy for incur-
mal tumor FDG uptake; activity in the tumor able NSCLC. Use of SUV-based assessment follow-
absent or similar to mediastinum. ing radiotherapy may be compromised by a number of
(2) PMR (partial metabolic response)—any appre- factors, including the commonly observed and
ciable reduction in intensity of tumor FDG uptake sometimes intense inflammatory reaction to radio-
or tumor volume. No disease progression at other therapy in normal tissues. These changes may have a
sites. measured SUV in the ‘‘malignant’’ range but can
(3) SMD (stable metabolic disease)—no appreciable usually be identified as such on qualitative assess-
change in intensity of tumor FDG uptake or ment. Post-radiotherapy changes conform to the vol-
tumor volume. No new sites of disease. ume of aerated lung in the radiation treatment
(4) PMD (progressive metabolic disease)—apprecia- volume, are of a geographic rather than segmental or
ble increase in tumor FDG uptake or volume of anatomical distribution and are non-congruent with
known tumor sites or evidence of disease pro- the biodistribution of uptake in tumoral sites on
gression at other intrathoracic or distant meta- baseline scanning. Residual disease, on the other
static sites. hand, conforms to the position of initial tumor
Responses were correlated with subsequent sur- allowing for anatomical distortion relating to further
vival. Median survival after follow-up PET was collapse or re-expansion of lung parenchyma and
24 months. There was poor agreement between PET tends to maintain a lobular shape. Similarly, tumoral
and CT responses (weighted kappa = 0.35), which uptake tends to respect and follow natural tissue
were identical in only 40% of patients. An example barriers such as the pleura of the oblique fissure
of PET response after radical chemoradiation is whereas radiation changes are not influenced by such
shown in Fig. 2. There were significantly more boundaries. The presence of FDG uptake in previ-
complete responders on PET (n = 34) than CT ously normal lung after radiotherapy has a strong
(n = 10) while fewer patients were judged to be correlation with the presence of radiation pneumonitis
non-responders (12 vs. 20) or non-evaluable (0 vs. (Mac Manus et al. 2010).
6) by PET. Both CT and PET responses were Thus, PET appears to be far superior to CT scan-
individually significantly associated with survival ning for predicting survival after radical radiation
duration, but on multifactor analysis including the therapy. The powerful prognostic information avail-
known prognostic factors CT response, performance able from post-treatment PET may encourage the
status, weight loss and stage, only PET metabolic development of investigational ‘‘response-adapted’’
response was significantly associated with survival therapeutic approaches. It also raises the possibility
duration (p \ 0.0001). that early PET response, during a course of radio-
The best method for response assessment after therapy could provide early prognostic information
therapy has not yet been determined (Wahl et al. and provide a basis for modifying therapy during the
2009), whether visual or quantitative approaches. treatment course.
PET and PET/CT in Treatment Planning 181

Fig. 2 Coronal images at


baseline FDG PET/CT before
(left upper panel) and after
chemoradiotherapy (post-
CRT, right upper panel)
reveal high activity in the
mid-zone of the left lung.
Evaluation of the sagittal
projection images (middle
panels) clearly demonstrate
that the site of the primary
tumour (blue arrow) losing its
metabolic signal following
treatment whereas new
abnormality with sharply
demarcated superior and
inferior borders (red arrows),
conforming to the cranio-
caudal extent of the radiation
treatment volume, became
apparent. The fused FDG
PET/CT images (lower
panels) confirmed loss of
metabolic abnormality (blue
arrows) in the primary lesion
between the baseline (left) and
post-treatment (right) scans,
which also revealed a sharply
demarcated abnormality (red
arrows) crossing the oblique
fissure and corresponding the
lateral margin of the treatment
volume

false positives related to inflammatory conditions.


7 The Future of PET in NSCLC The excellent clinical performance of FDG PET
suggests that this is not a major practical limitation in
7.1 Use of PET Tracers Other Than FDG countries with a relatively low prevalence of granu-
in Staging and Treatment Response lomatous lung disease. Even where diseases such as
Assessment in Lung Cancer tuberculosis and histoplasmosis are more common,
the combination of the intensity and pattern of uptake,
One of the theoretical limitations of FDG as a tracer combined with consideration of the pre-test proba-
for the evaluation of lung cancer is the presence of bility of disease enables many potential false positive
182 M. P. Mac Manus and R. J. Hicks

results based purely on the intensity of uptake (SUV) FDG had similar diagnostic performance although
to be prospectively identified. Even when this cannot FDG yielded significantly higher SUV’s than
11
be achieved, most conditions that cause false positive C-methionine.
results represent important pathologies that benefit Enhanced production of cell membranes in cancer
from specific diagnosis and therapy. Nevertheless, cells requires uptake of choline to form phosphatidyl-
there has been interest in assessing alternative tracers choline. Accordingly, radiolabeled choline analogs
for tumor imaging that may be less prone to uptake in have been investigated as a potential cancer imaging
inflammatory diseases. Early studies included com- agents. Initial studies focused on 11C-choline and
parison between FDG PET and 201Tl, a tumor imag- involved comparison with FDG PET in 29 patients with
ing agent commonly used in conventional nuclear biopsy proven NSCLC. This study demonstrated
medicine. The hope that 201Tl SPECT might prove superior sensitivity of 11C-choline for detection of
more specific than FDG was not realized. The lower mediastinal nodes. These results were not however
spatial and contrast resolution generally achieved confirmed by a subsequent study from the Netherlands,
with SPECT limits its ability to detect disease in non- which found that this tracer has lower sensitivity
enlarged mediastinal nodes and beyond the thorax, for mediastinal nodal involvement. An important
which, as shown above, accounts for the major observation in this study was the ability of 11C-choline
incremental value of FDG PET compared to CT and to detect brain metastases that are not seen on FDG PET
much of its clinical impact. due to high uptake in normal cerebral structures. This
Recognizing the instrumentation advantages of observation is pertinent to other tracers that also have
PET, comparison with other PET tracers is probably low accumulation in the brain, including 11C-methio-
more important. One of the first PET agents to be nine. Fluorinated choline analogs have recently been
compared was the amino acid tracer, 11C-methionine. described. Preliminary studies of one of these tracers
In a study looking at the accuracy of 11C-methionine for evaluation of lung cancer were, however, not
for mediastinal nodal staging, superior accuracy encouraging (Pauleit et al. 2005).
compared to CT was demonstrated using histopa- Another potential imaging target of lung cancer cells
thological validation with results comparable to those is their high proliferation. Proliferative rate may also
reported using FDG PET. However, this study did not provide insights into the biological activity and prog-
directly compare the 11C-methionine results with nosis of lung cancers. Although there does appear to be
those from FDG PET. In a small series from Sweden a relationship between FDG uptake and proliferation in
that did compare FDG and 11C-methionine, all pri- NSCLC (Higashi et al. 2000), there are factors other
mary tumors were equally well visualized by both than proliferation that potentially drive FDG uptake in
tracers and since there were no false positive FDG cancer cells. One of these factors is hypoxia, which
results, the possibility that amino acid imaging may increases expression of glucose transporters and
have a lower propensity for false positive results glycolytic enzymes but decreases cell proliferation.
could not be evaluated. A larger study from Japan Hence, tracers that more directly reflect cell prolifera-
found that the performance of both tracers was similar tion, such as tracers of DNA synthesis are attractive.
with a marginally higher specificity and accuracy with Various thymidine analogs have been developed for
11
C methionine but did not reach statistical signifi- PET imaging. These include 11C-thymidine and the
cance (Sasaki et al. 2001). A more recent, but rela- fluorinated analog FLT. Studies in lung tumors have
tively small study evaluating 15 solitary pulmonary demonstrated the feasibility of FLT for evaluating cell
nodules suggested that 11C methionine may be both proliferation but this tracer does not appear to be
more sensitive for some malignancies, such as gastric superior to FDG for lung cancer staging.
and thyroid cancer, known to sometimes have rela- A number of studies are now evaluating the utility
tively low FDG-avidity and more specific with lack of of FLT for therapeutic monitoring of radiotherapy and
significant uptake into inflammatory lesions, such as molecular targeted therapies. Although promising for
tuberculosis. In the restaging setting, where inflam- the evaluation of molecular targeted agents that may
matory changes may pose difficulties in determining lead to cell stasis rather than death, a preliminary
nature of increased FDG activity, the MD Anderson study comparing early FDG and FLT response sug-
Cancer Center group found that 11C-methionine and gested that both were predictive of progression-free
PET and PET/CT in Treatment Planning 183

survival but only early (day 14) FDG response pre- an extremely complex undertaking. Efforts to develop
dicted overall survival (Mileshkin et al. 2011). These methodology for respiratory gated PET have been
results have also been confirmed in a study from reported (Nehmeh et al. 2002) and offer the potential
Germany. for highly sophisticated treatment planning and
The ability of FLT to assess the extent and activity delivery (Wurm et al. 2006). Whether the resource
of bone marrow could provide a useful diagnostic tool implications of such an approach make it practical
in assessing the risk of radiotherapy in heavily pre- and affordable for routine clinical application remains
treated patients with recurrent disease. It has also to be seen but for patients in whom lung function is
recently been used to understand the effects of novel marginal for radical therapy, these highly targeted
forms of radiotherapy, such as carbon ion therapy, on approaches may be critical to outcome.
bone marrow function (Koizumi et al. 2011).

7.3 Response Adapted Radiotherapy:


7.2 Respiratory Gating of PET Data Targeting Tumor Subvolumes
During a Course of Radiotherapy
The acquisition of the emission data used to recon-
struct PET images occurs over several minutes and One of the most promising areas in radiotherapy
therefore integrates the effects of respiratory move- research is the use of PET to study changes that occur
ment. This has the effect of increasing the apparent in the tumor during a course of therapy. We know that
supero-inferior size of lesions near the base of the FDG PET is superior to CT for definitive assessment
lungs that move predominantly in the coronal plane of treatment response after chemoradiotherapy for
during respiration and the antero-posterior dimensions NSCLC but tumor imaging during therapy may allow
of lesions in the anterior aspect of the lungs that move for modification of the treatment course in real time.
mainly in the axial plane. In both circumstances, Different regions of the target contain different den-
apparent activity in slightly reduced by this move- sities of tumor cells and these densities change during
ment due to partial volume effects. CT scanning, therapy. Serial PET imaging during a course of RT
however, is acquired very rapidly and multi-slice could potentially provide prognostic information and
scanners can acquire images sufficiently fast to more importantly could provide an opportunity to
effectively ‘‘freeze’’ respiratory motion. Alternatively, give a higher RT dose to regions of tumor that show
CT scan images can be acquired during breath hold- the poorest metabolic or proliferative response during
ing at a given phase of respiration. Being derived therapy. Kong et al. at University of Michigan studied
from instantaneous images of the relative position of the correlation between FDG PET scans performed
organs, the location of lesions on CT planning images during RT with the definitive response on a post
does not necessarily correspond to the averaged or treatment PET scan several months later. FDG-PET/
integrated position of lesions detected by emission CT scans were acquired before, during, and after RT.
scanning. It is clear that respiratory movement can Tumor and lung metabolic responses were assessed
lead to misregistration of PET and CT lesions on qualitatively by physicians and quantitatively by
fused PET and CT images, whether acquired on stand normalized peak FDG activity. Of 15 patients, 11 had
alone or combined devices. Although this is not a PMR, two patients had CMR, and two patients had
particularly frequent diagnostic problem, it may pose stable disease at approximately 45 Gy during RT. The
difficulties when determining the GTV and PTV for qualitative metabolic response during RT correlated
radiotherapy. The approach at Peter MacCallum with the overall response post-RT (p = 0.03). At our
Cancer Centre has been to assume that, since the PET own centre we are exploring PET imaging with both
data represents the integrated position throughout the FDG and the proliferation marker 18F-30 -deoxy-30 -
respiratory cycle and the average position likely to fluoro-l-thymidine (FLT) to assess tumor response
also apply during a radiotherapy treatment episode, during therapy. Pilot data show that significant
PET should be used to plan the GTV. An alternative reductions in tumor and bone marrow FLT uptake
approach would be to perform respiratory gating of occur during radiotherapy (Everitt et al. 2009). Tumor
both the PET, CT and radiotherapy delivery. This is responses vary widely between patients but it is too
184 M. P. Mac Manus and R. J. Hicks

early to determine how these interim responses are Bayne M et al (2010) Reproducibility of ‘‘intelligent’’ con-
correlated with eventual outcome and too early to touring of gross tumor volume in non-small-cell lung cancer
on PET/CT images using a standardized visual method. Int J
employ them in modifying target volumes outside of a Radiat Oncol Biol Phys 77:1151–7
clinical trial. Biehl KJ et al (2006) 18F-FDG PET definition of gross tumor
The availability of PET tracers that can evaluate volume for radiotherapy of non-small cell lung cancer: is a
biological processes relevant to radiotherapy offers single standardized uptake value threshold approach appro-
priate? J Nucl Med 47:1808–1812
the prospect of biologically-adapted radiotherapy Binns DS et al (2011) Compliance with PET acquisition
(Gregoire et al. 2007). For example, dose-painting of protocols for therapeutic monitoring of erlotinib therapy in
regions of hypoxia could be facilitated by PET imaging an international trial for patients with non-small cell lung
with a range of tracers that are suitable for detection of cancer. Eur J Nucl Med Mol Imaging 38:642–650
Blum R et al (2004) Impact of positron emission tomography
hypoxia (Reischl et al. 2007). Radiolabeled mono- on the management of patients with small-cell lung cancer:
clonal antibodies, such as 89Zr-bevacizumab, may preliminary experience. Am J Clin Oncol 27:164–171
help to identify patients who could benefit from Bowden P et al (2002) Measurement of lung tumor volumes
inclusion of such agents into combination therapies using three-dimensional computer planning software. Int J
Radiat Oncol Biol Phys 53:566–573
with radiotherapy. Bradley JD et al (2004) Positron emission tomography in
limited-stage small-cell lung cancer: a prospective study.
J Clin Oncol 22:3248–3254
8 Conclusions Buck AK et al (2010) Economic evaluation of PET and PET/
CT in oncology: evidence and methodologic approaches.
J Nucl Med Technol 38:6–17
PET scanning is vastly superior to conventional Caldwell CB et al (2001) Observer variation in contouring
methods used in staging lung cancer and with the gross tumor volume in patients with poorly defined non-
widespread adoption of PET/CT this superiority has small-cell lung tumors on CT: the impact of 18FDG-hybrid
PET fusion. Int J Radiat Oncol Biol Phys 51:923–931
only increased. PET/CT should now be considered a Dahele M et al (2008) Developing a methodology for three-
standard tool for radiotherapy planning in NSCLC dimensional correlation of PET-CT images and whole-
because it provides the most accurate estimate of the mount histopathology in non-small-cell lung cancer. Curr
true disease extent currently available. The use of Oncol 15:62–69
Deniaud-Alexandre E et al (2005) Impact of computed
PET to exclude patients with extensive and, currently, tomography and 18F-deoxyglucose coincidence detection
generally incurable disease from potentially toxic emission tomography image fusion for optimization of
radical radiotherapy will significantly improve the conformal radiotherapy in non-small-cell lung cancer. Int J
overall results of treatment with this modality and Radiat Oncol Biol Phys 63:1432–1441
De Ruysscher D et al (2005) Effects of radiotherapy planning
early diagnosis of limited recurrent disease could with a dedicated combined PET-CT-simulator of patients
potentially facilitate salvage therapies. Furthermore, with non-small cell lung cancer on dose limiting normal
by decreasing futile attempts at curative treatment, tissues and radiation dose-escalation: a planning study.
expensive radical radiotherapy resources can be more Radiother Oncol 77:5–10
Dunagan D et al (2001) Staging by positron emission tomog-
effectively used. The evidence base for PET and PET/ raphy predicts survival in patients with non-small cell lung
CT for staging and radiotherapy planning in NSCLC cancer. Chest 119:333–339
is becoming so compelling that it is difficult to Eschmann SM et al (2007) Impact of staging with 18F-FDG-
imagine treating a patient with radical radiotherapy PET on outcome of patients with stage III non-small cell
lung cancer: PET identifies potential survivors. Eur J Nucl
without access to this technology. Nevertheless, fur- Med Mol Imaging 34:54–59
ther study is necessary to define the cost effectiveness Everitt S et al (2009) Imaging cellular proliferation during
of this very useful but expensive modality. chemo-radiotherapy: a pilot study of serial 18F-FLT
positron emission tomography/computed tomography imag-
ing for non-small-cell lung cancer. Int J Radiat Oncol Biol
Phys 75:1098–1104
References Everitt S et al (2010) High rates of tumor growth and disease
progression detected on serial pretreatment fluorodeoxyglu-
cose-positron emission tomography/computed tomography
Ball D, Smith J, Wirth A, Mac Manus M (2002) Failure of T scans in radical radiotherapy candidates with nonsmall cell
stage to predict survival in patients with non-small-cell lung lung cancer. Cancer 116:5030–5037
cancer treated by radiotherapy with or without concomitant Gould MK et al (2003) Test performance of positron emission
chemotherapy. Int J Radiat Oncol Biol Phys 54:1007–1013 tomography and computed tomography for mediastinal
PET and PET/CT in Treatment Planning 185

staging in patients with non-small-cell lung cancer: a meta- survival correlates with metastatic disease burden. Acta
analysis. Ann Intern Med 139:879–892 Oncol 42:48–54
Gregoire V, Haustermans K, Geets X, Roels S, Lonneux M MacManus M et al (2009) Use of PET and PET/CT for
(2007) PET-based treatment planning in radiotherapy: a radiation therapy planning: IAEA expert report 2006–2007.
new standard? J Nucl Med 48(Suppl 1):68S–77S Radiother Oncol 91:85–94
Hicks RJ (2009) Role of 18F-FDG PET in assessment of Mah K et al (2002) The impact of (18)FDG-PET on target and
response in non-small cell lung cancer. J Nucl Med critical organs in CT-based treatment planning of patients
50(Suppl 1):31S–42S with poorly defined non-small-cell lung carcinoma: a
Hicks RJ, Mac Manus MP (2003) 18F-FDG PET in candidates prospective study. Int J Radiat Oncol Biol Phys 52:339–350
for radiation therapy: is it important and how do we validate Mileshkin L et al (2011) Changes in 18F-fluorodeoxyglucose
its impact? J Nucl Med 44:30–32 and 18F-fluorodeoxythymidine position emission tomogra-
Hicks RJ et al (2001) (18)F-FDG PET provides high-impact phy imaging in patients with non-small cell lung cancer
and powerful prognostic stratification in staging newly treated with erlotinib. Clin Cancer Res 17:3304–3315
diagnosed non-small cell lung cancer. J Nucl Med 42: Munley MT et al (1999) Multimodality nuclear medicine
1596–1604 imaging in three-dimensional radiation treatment planning
Higashi K et al (2000) FDG PET measurement of the for lung cancer: challenges and prospects. Lung Cancer
proliferative potential of non-small cell lung cancer. 23:105–114
J Nucl Med 41:85–92 Nehmeh SA et al (2002) Effect of respiratory gating on
Hong R, Halama J, Bova D, Sethi A, Emami B (2007) quantifying PET images of lung cancer. J Nucl Med
Correlation of PET standard uptake value and CT window- 43:876–881
level thresholds for target delineation in CT-based radiation Nestle U et al (2006) Target volume definition for (18)F-FDG
treatment planning. Int J Radiat Oncol Biol Phys 67: PET-positive lymph nodes in radiotherapy of patients with
720–726 non-small cell lung cancer. Eur J Nucl Med Mol Imaging
Jaffe CC (2006) Measures of response: RECIST, WHO, and 33:263–269
new alternatives. J Clin Oncol 24:3245–3251 Nestle U et al (1999) 18F-deoxyglucose positron emission
Koizumi M et al (2011) Uptake decrease of proliferative PET tomography (FDG-PET) for the planning of radiotherapy in
tracer FLT in bone marrow after carbon ion therapy in lung lung cancer: high impact in patients with atelectasis. Int J
cancer. Mol Imaging Biol 13:577–582 Radiat Oncol Biol Phys 44:593–597
Kolodziejczyk M et al (2011) Impact of [(18)F]Fluorodeoxyglu- Nestle U et al (2005) Comparison of different methods for
cose PET-CT staging on treatment planning in radiotherapy delineation of 18F-FDG PET-positive tissue for target
incorporating elective nodal irradiation for non-small-cell volume definition in radiotherapy of patients with non-
lung cancer: a prospective study. Int J Radiat Oncol Biol Phys Small cell lung cancer. J Nucl Med 46:1342–1348
80(4):1008–1114 Pauleit D et al (2005) PET with O-(2–18F-Fluoroethyl)-L-
Lever AM, Henderson D, Ellis DA, Corris PA, Gilmartin JJ Tyrosine in peripheral tumors: first clinical results. J Nucl
(1984) Radiation fibrosis mimicking local recurrence in Med 46:411–416
small cell carcinoma of the bronchus. Br J Radiol Pommier P et al (2010) Impact of (18)F-FDG PET on treatment
57:178–180 strategy and 3D radiotherapy planning in non-small cell
Mac Manus MP et al (2010) Association between pulmonary lung cancer: A prospective multicenter study. AJR Am J
uptake of fluorodeoxyglucose detected by positron emission Roentgenol 195:350–355
tomography scanning after radiation therapy for non-small- Reischl G et al (2007) Imaging of tumor hypoxia with
cell lung cancer and radiation pneumonitis. Int J Radiat [124I]IAZA in comparison with [18F]FMISO and
Oncol Biol Phys 80:1365–1371 [18F]FAZA–first small animal PET results. J Pharm Pharm
Mac Manus MP et al (2001) F-18 fluorodeoxyglucose positron Sci 10:203–211
emission tomography staging in radical radiotherapy can- Sasaki M et al (2001) Comparison of MET-PET and FDG-PET
didates with nonsmall cell lung carcinoma: powerful for differentiation between benign lesions and malignant
correlation with survival and high impact on treatment. tumors of the lung. Ann Nucl Med 15:425–431
Cancer 92:886–895 Stroobants S, Verschakelen J, Vansteenkiste J (2003) Value of
Mac Manus MP et al (2002) Early mortality after radical FDG-PET in the management of non-small cell lung cancer.
radiotherapy for non-small-cell lung cancer: comparison of Eur J Radiol 45:49–59
PET-staged and conventionally staged cohorts treated at a Toloza EM, Harpole L, Detterbeck F, McCrory DC (2003)
large tertiary referral center. Int J Radiat Oncol Biol Phys Invasive staging of non-small cell lung cancer: a review of
52:351–361 the current evidence. Chest 123:157S–166S
Mac Manus MP et al (2003) Positron emission tomography is Townsend DW, Beyer T (2002) A combined PET/CT scanner:
superior to computed tomography scanning for response- the path to true image fusion. Br J Radiol 75(Spec No):S24–
assessment after radical radiotherapy or chemoradiotherapy S30
in patients with non-small-cell lung cancer. J Clin Oncol van Baardwijk A et al (2007) PET-CT-based auto-contouring in
21:1285–1292 non-small-cell lung cancer correlates with pathology and
MacManus MR et al (2003) FDG-PET-detected extracranial reduces interobserver variability in the delineation of the
metastasis in patients with non-small cell lung cancer primary tumor and involved nodal volumes. Int J Radiat
undergoing staging for surgery or radical radiotherapy— Oncol Biol Phys 68:771–778
186 M. P. Mac Manus and R. J. Hicks

van de Steene J et al (2002) Definition of gross tumor volume in node staging on the radiation treatment volumes in patients
lung cancer: inter-observer variability. Radiother Oncol with non-small cell lung cancer. Radiother Oncol 55:
62:37–49 317–324
van Der Wel A et al (2005) Increased therapeutic ratio by Wahl RL, Jacene H, Kasamon Y, Lodge MA (2009) From
18FDG-PET CT planning in patients with clinical CT stage RECIST to PERCIST: Evolving Considerations for PET
N2-N3M0 non-small-cell lung cancer: a modeling study. response criteria in solid tumors. J Nucl Med 50(suppl
Int J Radiat Oncol Biol Phys 61:649–655 1):122S–150S
van Loon J et al (2008) 18FDG-PET based radiation planning Wurm RE et al (2006) Image guided respiratory gated hypofrac-
of mediastinal lymph nodes in limited disease small cell tionated stereotactic body radiation therapy (H-SBRT) for
lung cancer changes radiotherapy fields: a planning study. liver and lung tumors: initial experience. Acta Oncol
Radiother Oncol 87:49–54 45:881–889
Vanuytsel LJ et al (2000) The impact of (18)F-fluoro-2-deoxy- Yaremko B et al (2005) Thresholding in PET images of static
D-glucose positron emission tomography (FDG-PET) lymph and moving targets. Phys Med Biol 50:5969–5982
Target Volume Definition in Non-Small Cell
Lung Cancer
Lucyna Kepka and Milena Kolodziejczyk

Contents Abstract
Proper target volume delineation is a crucial stage
1 Introduction.............................................................. 187 of treatment planning, so any error introduced in
2 Gross Tumor Volume Definition ........................... 188 this process is a systematic error and cannot be
2.1 GTV for Tumor ......................................................... 188 quantified and/or detected by modern treatment
2.2 GTV for Nodes.......................................................... 189 technologies, unlike other sources of geometrical
2.3 Problems Related to the Use
uncertainties. All steps of target definition should
of PET-CT for GTV Definition ................................ 189
2.4 Definition of GTV After Chemotherapy .................. 190 be standardized. In non-small cell lung cancer
radiotherapy, there are specific problems related to
3 Clinical Target Volume Definition ........................ 191
3.1 Microscopic Spread Around Primary Tumor the definition of all three consecutive target
and Pathologic Lymph Nodes................................... 191 volumes recommended by ICRU: gross tumor
3.2 Elective Nodal Irradiation ......................................... 192 volume (GTV), clinical target volume (CTV), and
3.3 Lymph Node Stations Delineation for ENI Issues... 193 planning target volume (PTV). In GTV delinea-
3.4 Postoperative Radiotherapy Target Volume............. 194
tion, the proper imaging, e.g., standardized way of
4 Planning Target Volume Definition ...................... 196 the use of CT and PET-CT, and continuous
4.1 Internal Target Volume: Respiratory Motion
radiological training of radiation oncologists are
Management............................................................... 196
4.2 Margins for Set-up .................................................... 197 emphasized. For CTV, we still lack robust data on
the margin which is necessary to expand around
References.......................................................................... 197
GTV of the tumor and pathologic lymph nodes to
adequately account for microscopic spread. Addi-
tionally, elective nodal irradiation is still a source
of controversies. For PTV definition, major
increase in technologies is involved. It leads in
some cases to improvement of the tumor coverage
and sparing of organs at risk, but as this process is
expensive and time consuming, it might not be
always beneficial.

1 Introduction
L. Kepka (&)  M. Kolodziejczyk
Target volume delineation is the most important step
M. Sklodowska-Curie Memorial Cancer and Institute
of Oncology, Warsaw, Poland in the radiation therapy planning. While set-up and
e-mail: lucynak@coi.waw.pl respiratory motion displacements can be quantified

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 187
DOI: 10.1007/174_2011_231,  Springer-Verlag Berlin Heidelberg 2011
188 L. Kepka and M. Kolodziejczyk

Table 1 The International Commission on Radiation Units


and Measurements (ICRU 1993, 1999) definitions of target 2 Gross Tumor Volume Definition
volumes
Gross tumor Macroscopic extent of the malignant All the steps of gross tumor volume (GTV) definition
volume (GTV) growth, e.g., clinically palpable and/or
visualized by the imaging should be standardized. Inter-observer variability in
the contouring of the GTV is a major source of the
Clinical target Anatomical-clinical concept that needs
volume (CTV) to be defined before delineation. It uncertainty in radiotherapy planning. Reported dif-
contains GTV and/or subclinical ferences in the contouring varied between 12 and 60%
microscopic disease which should be on average (Bowden et al. 2002; van de Steene et al.
eliminated
2002; Giraud et al. 2002; Steenbakkers et al. 2005).
Planning target Geometrical concept. A 3D-expansion The numerous measures to overcome this problem are
volume (PTV) of the CTV to account for all
geometrical uncertainties (for target recommended. The first one is the formulation and
and organ at risk motion, set-up error, obedience of the departmental contouring guidelines.
delineation, anatomical changes during The definition of appropriate windows setting for
treatment) computerized tomography (CT), appropriate training
Internal target A component of PTV specified by in radiology and cooperation with radiologist, and the
volume (ITV) Report 62 (ICRU 1999), e.g.,
use of information from PET-CT (in a standardized
CTV ? margin for motion
way) are also related to better reproducibility of the
delineated targets (Bowden et al. 2002; Steenbakkers
et al. 2005, 2006; Greco et al. 2007).
and corrected at the individualized basis with the
modern technologies, target contouring is still prone
to the risk of introducing human error. The important 2.1 GTV for Tumor
inter-observer variability in the target definition, also
for lung cancer, has been documented extensively In the era of the three-dimensional conformal radio-
(Bowden et al. 2002; Giraud et al. 2002; Steenbakkers therapy (3D-CRT), the definition of the GTV is based
et al. 2005, 2006; Kepka et al. 2007). The error on the CT. The CT performed for planning purposes
introduced in the target definition is a systematic error is executed in the treatment position; the consider-
so it impacts the tumor coverage heavily (van Herk ation of the respiratory motion is crucial for radio-
et al. 2000). Consequently, a continuous education therapy planning. The CT is not done at the breath
and training of the radiation oncologists in this field is hold as for diagnostic purposes. Slow scanning which
needed. allows for consideration of the tumor movement in
In this chapter, the issues pertinent to the three different breathing phases leads to the image blurring.
stages of the target volume definition in radiotherapy Currently, the 4D-imaging prevents this inconve-
of non-small cell lung cancer (NSCLC) will be dis- nience. It records a respiratory signal during image
cussed. Those three stages of target volume definition acquisition and enables retrospective reconstruction
follow the recommendation of the International of a dataset from multiple phases of respiratory cycle.
Commission on Radiation Units (ICRU) and Mea- A choice of appropriate window setting for delinea-
surements Report 50 (ICRU 1993) and Report 62 tion is of crucial value. For delineation of tumors
(ICRU 1999). The ICRU recommends the consecu- located within pulmonary tissue, it is recommended to
tive definition of the gross tumor volume (GTV), use so-called ‘‘pulmonary window’’ setting. The best
clinical target volume (CTV), and Planning Target concordance between the radiological image and the
Volume (PTV). Report 62 recommends additional actual dimensions of the parenchymal tumor has been
definition within PTV called the Internal Target established at the window width 1,600 and level
Volume (ITV). The ICRU definitions of respective -600. For tumors located centrally, it is useful to use
target volumes are summarized in Table 1. Thus the also mediastinal window setting with recommended
specific issues related to the target definition in window width of 400 and level of 20 (de Ruysscher
NSCLC radiotherapy will be presented in the light of et al. 2010). However, the ‘‘in house’’ measurements
the ICRU recommendations. of appropriate window setting should be done in each
Target Volume Definition in Non-Small Cell Lung Cancer 189

department, as calibration of CT may differ between pathologically excluded) and the FDG-PET negative
centers. The contrast injection for planning CT may lymph nodes with a diameter that exceeds 1.5 cm in
be useful in case of tumors localized in the hilar the nodal GTV. However, there are also more strict
region for the purpose of distinguishing of vessels in guidelines for definition of the nodal GTV which
this region. were traditionally used within Radiation Therapy
For central locations of tumor with endobronchial Oncology Group (RTOG). According to those
component, the bronchoscopy findings should be guidelines, all FDG-PET positive lymph nodes and all
considered, because even high resolution CT does not lymph nodes with a diameter in short axis [1.0 cm
visualize this component and the FDG-PET sensitiv- were contoured as nodal GTV (Bradley et al. 2010).
ity and specificity is estimated at 73 and 85%, Lymph nodes should be contoured on the mediastinal
respectively (Pasic et al. 2005). The special clinical window as defined above for central components of
case for such setting represents a roentgenographi- tumors.
cally occult carcinoma, a rare entity treated usually
with combination of external-beam and endobron-
chial brachytherapy. Currently, we do not have clear
guidelines for GTV delineation in those cases, 2.3 Problems Related to the Use
because series reported so far started with CTV for of PET-CT for GTV Definition
delineation which may represent a reasonable solution
in case of the poor tumor visualization (Saito et al. The value of the use of PET-CT for radiotherapy of
2000). NSCLC is described in detail in another chapter of
this book. Only some specific problems for GTV
definition will be overviewed in this paragraph. The
2.2 GTV for Nodes overview of studies on treatment planning of lung
cancer with PET-CT showed 5–64 and 0–70% of
This component of GTV is delineated on the CT increase and decrease of radiation volumes defined in
scans; however, the PET-CT is currently quite rou- the CT alone, respectively (Macmanus et al. 2009).
tinely used for diagnosis of metastases of NSCLC to For the nodal GTV, those changes are related to
the mediastinal and hilar lymph nodes. It is due to the higher accuracy of PET-CT than CT alone in the
higher accuracy of the mediastinal staging in PET mediastinal staging discussed above. For the tumor, it
than in CT. In an overview done by Silvestri et al. is mainly related to the superior value of the PET in
(2007), the PET scanning sensitivity and specificity the distinction of the tumor from atelectasis (Nestle
for mediastinal staging was 0.74 and 0.85 compared et al. 2006). However, we should be cautious of the
to respective values of 0.51 and 0.85 for CT scanning. uncritical interpretation of those changes that result
Thus the PET-CT improves the mediastinal staging in from PET use. The outcomes of those studies may
comparison with CT alone, however, it is far from differ because of the variability in methodology
being perfect. The findings from both examinations across centers. Therefore, to unify PET-CT scan
should be considered carefully in the definition of procedure in treatment planning, the European
the nodal GTV. If radiotherapy planning was based Association of Nuclear Medicine (EANM) published
on the CT only, the usual policy would be to include the guidelines for tumor PET imaging (Boellard et al.
in the GTV, all mediastinal lymph nodes with a 2010). The recommendations for the use of PET-CT
diameter at short axis of higher than 1.0 cm. With the in lung cancer radiotherapy were also recently pub-
use of the PET-CT, we know that for 1.0–1.5 cm lished (de Ruysscher et al. 2010). The GTV generated
FDG-PET negative lymph nodes, the probability of from functional imaging with poor image resolution
metastatic involvement is below 5%. However, for cannot be surrogate for a definition of the GTV
FDG-PET negative lymph nodes measuring in short defined by the CT which is able to better visualize the
axis [1.5 cm, the probability of metastatic involve- tumor. It is only an additional source of information
ment is 21% (de Langen et al. 2006). So the formu- which should be taken into account when contouring
lation of the guidelines may indicate that we should targets. For contouring, the key question is the
include all FDG-PET positive lymph nodes (unless method of outlining of the GTV on the FDG-PET
190 L. Kepka and M. Kolodziejczyk

based images. The methods for outlining were used: Table 2 Phase III clinical studies which had in one of arms
(1) the qualitative visual method, (2) the GTV 2.5 radiotherapy delivered after induction chemotherapy and rec-
ommended the use of pre-chemotherapy volumes for target
standardized uptake value (SUV) units, (3) the linear definition
adaptive SUV threshold function method, and (4) the
Trial, year Design of the study
use of any threshold of local maximal uptake value in
which the threshold of 41–42% established in the Le Chevalier Radiotherapy alone versus sequential
et al. 1991 radiochemotherapy
phantoms studies (Boellard et al. 2010; Black et al.
Dillman et al. Radiotherapy alone versus sequential
2004) is commonly used. Despite the fact that
1996 radiochemotherapy
thresholds of 42 (van Loon et al. 2010) and 50% (Wu
Furuse et al. Sequential radiochemotherapy versus
et al. 2010) were seen as fitting the best for the 1999 concurrent radiochemotherapy
diameter of the tumor reported on the pathology
Sause et al. 2000 Radiotherapy alone versus sequential
examination, the use of predefined thresholds is a radiochemotherapy
source of potential errors. It was proven that due to Curran et al. Sequential radiochemotherapy versus
the heterogeneity of the FDG uptake in the lung tumor 2003 concurrent radiochemotherapy
and poor spatial resolution of the PET leading to the Zatloukal et al. Sequential radiochemotherapy versus
partial volume effect, the automatic delineation with 2004 concurrent radiochemotherapy
any predefined threshold cannot provide the reliable Fournel et al. Sequential radiochemotherapy versus
GTV definition (Devic et al. 2010). The recom- 2005 concurrent radiochemotherapy
mended approach of contouring of the FDG-PET Huber et al. 2006 Induction chemotherapy followed by
images is the qualitative visual method (de Ruysscher radiotherapy or concurrent
et al. 2010). It is also not a perfect solution, as it radiochemotherapy
is related to the risk of inter-observer variability. Vokes et al. 2007 Concurrent radiochemotherapy versus
To overcome this problem, the contouring should be induction chemotherapy followed by
concurrent radiochemotherapy
held in close cooperation of the radiation oncologist
van Meerbeeck Induction chemotherapy followed by
and the nuclear medicine physician in experienced et al. 2007 surgery or definitive radiotherapy
centers.
Belderbos et al. Sequential radiochemotherapy versus
2007 concurrent radiochemotherapy
Thomas et al. Induction chemotherapy followed by
2.4 Definition of GTV After 2008 radiochemotherapy and surgery versus
Chemotherapy surgery and postoperative radiotherapy

When radiotherapy is used in case of tumor regression


after induction chemotherapy, the question is raised as
to which pre-, or post-chemotherapy volumes, should
be considered for GTV creation. All phase III clinical physicians working in the same institution (Tai et al.
studies (Table 2) which had in one of arms radio- 2004). It reflects a need for recommendations and
therapy delivered after induction chemotherapy rec- education on this very pertinent issue. The use of pre-
ommended the use of pre-chemotherapy volumes for chemotherapy volumes may be an additional source
target definition. Thus we should use a pre- of uncertainty and errors in radiotherapy planning,
chemotherapy volume for GTV definition except in especially, if only the visual inspection for compari-
cases where such approach would lead to an unac- son of images is used. The co-registration and fusion
ceptable risk of pneumonitis. Opposite approach may of pre- and post-chemotherapy imaging is a recom-
not be safe, because we lack evidence which indicates mended approach for adequate consideration of the
that its outcome in terms of local control is not initial disease status (Lagerwaard et al. 2002). In case
inferior. Despite a lack of evidence, the clinical of the use of PET-CT for radiotherapy planning, a
practice in this field varies considerably. Canadian minimum of 10 days should be taken after the last
patterns of care survey showed that 42% of radio- chemotherapy dose and planning or it might be even
therapists used post-chemotherapy target volumes and be better to do the examination as close to the next
the differences in this regard were seen even among chemotherapy as possible (Boellard et al. 2010).
Target Volume Definition in Non-Small Cell Lung Cancer 191

related to the presence of the microscopic spread


3 Clinical Target Volume Definition around pulmonary mass. Giraud et al. (2000) indi-
cated that adenocarcinoma histology requires larger
3.1 Microscopic Spread Around Primary CTV margins than squamous cell carcinoma. This
Tumor and Pathologic Lymph Nodes was not confirmed in a study done by van Loon et al.
(2010); however, a small sample group (34 cases)
The International Commission on Radiation Units would preclude such a finding. On the other hand,
(ICRU) and measurement reports 50 (ICRU 1993) larger GTV and higher tumor density on the CT scan
and 62 (ICRU 1999) recommend the addition of revealed to be the predictors of risk of the occurrence
margin to the GTV for microscopic disease extension of microscopic disease spread in this study. Of note,
to create the clinical target volume (CTV). There are neither the use of CT nor PET-CT had superior value
three distinct types of CTV margins in lung cancer in the visualization of the CTV and its covering.
radiotherapy, each one causing separate problems, Higher differentiation (lower grade) of adenocarci-
namely, margin within pulmonary parenchyma, mar- noma was related to significantly larger microscopic
gin for endobronchial spread, and margin for extra- disease extension (Grills et al. 2007); it was rather
capsular extension in mediastinal and hilar lymph related to the increased frequency of bronchioalveolar
nodes. Obviously, as it is a microscopic spread, we histology in lower grade tumors. Summarizing, the
have no imaging which can detect it. Our current CTV around GTV located within lung tissue is still a
knowledge on this issue is based on the correlation of source of uncertainty, as our knowledge on the pre-
imaging with pathologic data. diction of the risk of microscopic spread is very
For microscopic disease spread of NSCLC within limited. In the meantime, we should be aware of this
the pulmonary parenchyma, the seminal work was phenomenon and be particularly cautious about this
performed by Giraud et al. (2000) in which the margin if the radiation with sharp penumbra or tech-
pathologic findings were correlated with pre-opera- niques with rapid dose fall-off are used. The lung
tive CT images. It was found that for covering 95% of tumor should be defined on the pulmonary window,
microscopic disease around pulmonary GTV, the because its use led to the reduction (but not elimi-
margin of 8 and 6 mm is needed, respectively, for nation) of the potential error of omission of the CTV
adenocarcinoma and squamous cell carcinoma. The margin (Giraud et al. 2000; Grills et al. 2007; van
usual margin of 5 mm would cover 91% of squamous Loon et al. 2010).
cell carcinoma, but only in 80% of adenocarcinoma Endobronchial tumor spread may be seen as in-
cases. When the microscopic spread was evaluated for tramucosal, submucosal or intraparietal microscopic
35 patients with adenocarcinoma, the margin required tumor extension. This is largely discussed in surgical
for covering 90% of cases was 9 mm, if appropriate series and endobronchial brachytherapy. For surgical
pulmonary window was used for GTV definition specimen, the usual proximal safety margin from the
(Grills et al. 2007). Correlation of images with path- visible tumor on the bronchial level is 1.5–2.0 cm
ologic data may be biased by tissue deformations of (Kara et al. 2000). For external-beam radiotherapy,
resected lung. It has been shown that if the tissue we have no clear recommendations for use of margin
deformations are not taken into account, the under- for endobronchial microscopic tumor extension. It is
estimation of the microscopic disease extension in quite surprising, because we have data for central
vivo may occur up to factor of 2 (Stroom et al. 2007). tumors which shows that the proximal microscopic
van Loon et al. (2010) evaluated the CTV margins tumor extension occurs in 30% of cases and its mean
around GTV defined with CT alone or PET-CT with dimension is about 10 mm (Kara et al. 2001). In the
tissue deformations taken rigorously into account and absence of recommendations on the CTV contouring
they found that the necessary margin to cover 90% of within broncho-tracheal tract, it is wise to take this
cases was as large as 26 mm, even if 50% of 34 data into consideration for central tumors and addi-
evaluated specimens had no microscopic disease tionally incorporate the bronchoscopy findings into a
extension at all. It shows that the CTV margin still target definition.
represents the big uncertainty in the radiation oncol- Clinical target volume around involved lymph
ogy. For this reason, it is crucial to identify the factors nodes was not as extensively studied as the extent of
192 L. Kepka and M. Kolodziejczyk

margin around pulmonary tumor. In one study (Yuan randomized trial addressed this issue specifically
et al. 2007a) the margin of 3 mm for lymph nodes (Yuan et al. 2007b). In this trial, a total of 200 patients
was determined for lymph nodes of size up to 20 mm. were randomized between involved-field radiotherapy
However, for lymph nodes of 21–30 mm, the extra- to 68–74 Gy or ENI to 60–64 Gy. Patients in the
capsular microscopic extension might have reached involved-field arm achieved better overall response
up to 12 mm. Mediastinal lymph nodes larger than rate (90 vs. 79%, p = 0.032) and better 5-year local
30 mm were not included in the study. So this indi- control rate (51%) than those in the ENI arm (36%),
cates another source of uncertainty in the radiation p = 0.03. It resulted in the borderline significant
target planning. In the absence of the robust data on improvement of overall survival at two years
this issue, different measures are taken to overcome (p = 0.05), but not at five years. Since there were
such a potential source of errors. One of these is the differences in ENI use and in a total dose between two
inclusion of the whole lymph node station that con- arms, it remains unclear if the poorer outcome from
tains the involved lymph node(s) in the CTV ENI was due to the lower radiation dose used in the
(de Ruysscher et al. 2008; Kepka et al. 2009b). If such ENI arm or the use of ENI itself.
an approach is justified, the further studies are The opponents of ENI argue that, the low risk of
warranted. isolated nodal failure, below 10% in most retrospec-
tive studies does not justify the use of ENI (de
Ruysscher et al. 2010). The planning studies (Grills
3.2 Elective Nodal Irradiation et al. 2003) suggested a benefit of omission of ENI for
reduction of pulmonary and esophageal toxicity. This
Elective nodal irradiation (ENI) in lung cancer means was partially confirmed in the randomized study of
that CTV includes the volume of uninvolved areas of Yuan et al. (2007b), in which the patients receiving
hilum and mediastinum, and sometimes supraclavicu- involved fields concurrent with chemotherapy had
lar region, in view of eradication of probable less radiation pneumonitis (p = 0.04) than patients
micrometastases within this field. The ENI volume, as treated with concurrent radiochemotherapy with ENI.
a part of the CTV, remains an anatomic-clinical con- There was no difference for esophagitis. The retro-
cept. The mediastinal ENI volume could appear as a spective analysis by Fernandes et al. (2010) on 108
result of clinical compromise between the need of patients (60 receiving ENI and 48 treated with
eradication of subclinical disease and acceptable tox- involved fields) showed that ENI was related to an
icity. The diagnostic possibilities and technical facili- increased, but manageable esophageal toxicity
ties also contribute to the choice of the extent of ENI. (p = 0.04). It is also postulated that better staging
This concept has been historically changing. From the with PET-CT use allows for the use of more tailored
70s to the 90s, its position was growing which resulted fields. Additionally, we can argue that incidental
in the following recommendations ‘‘…for upper and irradiation (defined as radiation delivered outside of
middle lobe lesions, the mediastinal field should the defined targets volumes not intentionally, but just
include 5 cm below the carina and supra-clavicular as a result of beams configuration in areas of steeper
areas. For lower lobe lesions, the lower margin of the or more protracted dose fall-off) may also contribute
field is the diaphragm. Irradiation of contralateral to the sterilization of the potential micrometastases in
hilum remains controversial’’ (Rocmans et al. 1991). the neighboring lymph nodes.
However, advances in the technology which enabled All of opponent’s arguments are debatable. First of
better imaging, dose comparisons which indicated all, the so-called ‘‘low risk’’ of isolated nodal failures,
potential toxicity of ENI, and substantial evidence usually between 5 and 9%, does not seem to be really
about a low risk of isolated nodal failures defined as negligible. If only half of these recurrences would be
regional nodal failures outside radiation field led to the prevented by ENI, the survival improvement would
recommendations to abandon ENI, first in early stages be detectable (Kelsey et al. 2009). It is more than the
(Rowell and Williams 2001) and later on for stage III benefit from addition of chemotherapy to radiother-
disease (de Ruysscher et al. 2010). apy in NSCLC which changed the clinical practice.
However, the body of evidence for the use of only The toxicity of ENI has not been documented con-
involved field in NSCLC is weak, because only one vincingly enough up to now. PET-CT is more
Target Volume Definition in Non-Small Cell Lung Cancer 193

powerful tool than CT alone for detection of nodal delineation of lymph node stations (LNS) appears. In
metastases; however, it has no value for detection of the era of 3D-CRT, the recommendations for delinea-
micrometastases. It was recently demonstrated by tion of LNS using CT imaging were published as late as
Videtic et al. (2008) that more than 30% of stage III in 2005 (Chapet et al. 2005). This was related to the
NSCLC patients had nodal metastases in mediasti- controversies around ENI and its gradual abandon-
noscopy which were undetected in PET-CT. ment. However, there was a need for such guidelines,
Recently, the prospective study evaluated the value of for many purposes, out of which postoperative radio-
ENI in the reduction of risk of potential geographical therapy was one of the most pertinent. Those recom-
miss with radiotherapy planned without PET-CT. In mendations, known as the Atlas of Michigan, whose
75 patients for whom curative radiotherapy was name comes from the institution where the steps for
maintained after PET-CT, there were 20 cases of this important work had been taken, met with an urgent
potential (without pathologic confirmation) geo- need in lung cancer radiotherapy for providing the
graphical misses on the CT-alone based plans. Thir- guidelines allowing for the introduction of standards
teen patients from this group had ENI initially and generation of consensus in the delineation of
planned on CT-alone. For seven patients judged as uninvolved nodal areas. Atlas of Michigan provided
less advanced, the ENI was abandoned. It was dem- recommendations for delineation of hilar and medias-
onstrated that 90% isodose did cover entirely the tinal LNS on the CT scans, providing detailed
PET-based GTV only in 2 out of 13 patients with ENI description of borders of each LNS based on the
use. On the other hand, for 13 patients receiving ENI, structures visible on the CT axial images. It was based
the mean of minimum dose within missed GTV was on the N descriptors of the Mountain and Dressler
55% of prescribed total dose for gross disease while modification of the American Thoracic Society (MD-
for 7 patients without ENI it was 10%, p = 0.006. It ATS) LNS map (Mountain and Dressler 1997). This
suggests that if PET-CT is unavailable, ENI may to work needed clinical validation about reproducibility
some extent compensate for an inadequate dose of guidelines and feasibility of treatment plans done
coverage resulting from diagnostic uncertainties with nodal targets that were defined accordingly. In a
(Kołodziejczyk et al. 2010). study by Kepka et al. (2007), it was found that varia-
A recent review (Belderbos et al. 2008) concluded tions of delineation of LNS according to those guide-
that the use of ENI depends on many variables which lines were similar to all other studies on the variability
can be divided into tumor-, patient-, staging-, and in targets definition with a significant difference among
treatment-related. There is clinical situation, as, for physicians of about 36–60%. However, physicians
example, in advanced nodal stage when the use of working together in this field had fewer differences
ENI may be of value due to higher risk of isolated which indicate the need for a continuous training in the
nodal failure in radiologically uninvolved lymph proper use of those complex guidelines. This study also
nodes and demonstrated poorer sensitivity of PET- demonstrated that the strict following of these guide-
CT. In case of bulky mediastinal disease, the risk of lines in the setting of the slow scanning techniques led
occurrence of isolated nodal failure exceeded 20% to some pitfalls and difficulties in interpretation. Using
(Kepka et al. 2008). Another issue to be considered is this technique, the placement of the inferior limit of the
a poor staging (imaging) or radiotherapy used as the LNS 7 at the origin of the right middle lobe bronchus
only treatment modality for more advanced cases according to the Atlas led to the unusually distal
where toxicity added by ENI may be not so mean- delineation of this LNS, which was due to the relatively
ingful and potential for dose escalation is limited. large hilar mobility. The authors then modified this rule
to meet their needs and to meet the normal lung con-
straints limiting the delineation of LNS7 in the space
3.3 Lymph Node Stations Delineation between the right and left main bronchi. The users of
for ENI Issues the Atlas should also be aware that a strict following of
those guidelines in delineation of LNS with addition of
If one decides to use some form of ENI, regardless of appropriate margins according to the ICRU 62 rec-
purpose, whether for definitive radiotherapy or post- ommendations leads to the creation of the elective
operative radiotherapy (PORT), the issue of proper fields larger than traditionally defined in the 2D
194 L. Kepka and M. Kolodziejczyk

guidelines (Rocmans et al. 1991, RTOG 9410). This


phenomenon was demonstrated in the planning study
by Kepka et al. (2009a). One solution for this would be
to include fewer LNS in the elective field, e.g., only the
LNS with the highest probability of microscopic
invasion according to the histological type and location
(side and lobe) of the tumor. One of the propositions of
using such more selective ENI is software proposed by
Giraud et al. (2006) that evaluates the risk of medias-
tinal lymph node involvement from easily accessible
individual pretreatment parameters.
Recently, a new LNS map for the seventh edition of
TNM classification of lung cancer effective as of
January 1st, 2010 was published (Rusch et al. 2009).
This new LNS map is a consensus of the International
Association for the Study of Lung Cancer (IASLC)
experts and it reconciles differences among two LNS
maps and nomenclatures, the first one MD-ATS map
adopted by UICC (International Union against Cancer)
in the TNM classification and the Naruke map (Naruke
et al. 1978) used commonly in Japan. The borders of
LNS were specified in the way that allows users to
apply these definitions to clinical practice by CT, as the
appropriate CT illustrations were generated. There are
a number of changes for LNS delineation in compari-
son with MD-ATS map and consequently the Atlas of
Michigan guidelines provided for radiotherapists.
Figure 1 illustrates examples of differences in the LNS
delineation between Atlas of Michigan and a new map
proposed by IASLC. The most important changes
involve: description of supraclavicular and sternal
notch lymph nodes as level 1; shift of the left border of
level 2R and 4R from midline to the left wall of the
trachea; and precise description using anatomical
landmarks borders between LNS 4R and 10, 5 and 10,
10 and 11 bilaterally. Obviously, those differences
between two maps should be taken into account in Fig. 1 Difference in delineation of lymph node stations for
radiation oncology planning. The guidelines used for guidelines of Atlas of Michigan (Chapet et al. 2005) and
International Association of the Study for Lung Cancer
LNS delineation should be specified and reported in all (IASLC) staging committee of a new lymph node map for
treatment protocols involving the LNS delineation. 7th TNM classification of lung cancer (Rusch et al. 2009). a, b
Difference for lymph node stations 2R and 2L. It is a change in
the division between 2R and 2L (midline in the Atlas of
Michigan and left border of trachea in IASLC). a Delineation
3.4 Postoperative Radiotherapy Target according to the Atlas of Michigan (2R in red and 2L in
Volume orange). b Delineation according to the IASLC map (2R in blue
and 2L in green). c Difference in left hilar lymph node station—
Clinical target volume definition for postoperative 10L. In the IASLC map, 10L is delineated around main
bronchus (marked red in the picture), while in the Atlas of
radiotherapy (PORT) represents another source of Michigan, the medial border of 10L is the arbitrary imaginary
controversies, as no clear consensus and evidence line drawn between a lateral border of the pulmonary trunk and
exist in this field. It is related to the controversies with aorta descendens (marked light green in the picture)
Target Volume Definition in Non-Small Cell Lung Cancer 195

the use of PORT itself. The value of PORT for NSCLC


was questioned by the results of the PORT meta-anal-
ysis (1998) which included 2128 patients from nine
randomized trials in which PORT was compared with
surgery alone. This meta-analysis showed that there is
an increased relative risk of death of 21% with the use
of PORT. This deleterious effect was detected in
patients with pN0-1 disease. The detrimental effect of
PORT on survival was related to the excess of cardiac
and pulmonary deaths. No effect was detected in
patients with pN2 disease. It is now expected that for
the latter category, the benefit of PORT will be dis-
closed in the setting of use of adjuvant chemotherapy.
Improvement of survival with chemotherapy via
reduction or delay of distant metastases may lead to the Fig. 2 Target volume for postoperative radiotherapy (PORT)
representing a limited elective nodal irradiation—a coronal
disclosure of further benefit in overall survival by view. CTV (in green) for the right side includes bronchial
improvement of loco-regional control by the use of stump, ipsilateral hilar region, right/left paratracheal (4R/4L),
PORT. Such trials are ongoing or in preparation. and subcarinal lymph nodes. PTV (in red) represents
Recently, the experts from Lung Adjuvant Radio- CTV ? 1 cm margin
therapy Trial (Lung ART) Investigators Group that
prepared the randomized trial of PORT for pN2 NSCLC
patients demonstrated a large variability of CTV defi- (LNS 7), lower paratracheal lymph nodes (4R and 4L),
nition for PORT volumes up to threefold among experts 3A up to top of aortic arch, and LNS 5 (for left side), were
form EORTC. When the detailed protocol of CTV for included in the CTV. Decision about inclusion of ipsi-
PORT was provided, clear improvement was seen for lateral supraclavicular region in the CTV in case of
contouring of the lymph node stations (LNS) specified invasion of the lymph nodes above aortic arch was left at
by protocol. Only upper paratracheal lymph nodes (sta- the discretion of treating centres. For such defined CTV,
tions 2R and 2L according to Mountain and Dressler the margin of 1 cm was added to create PTV (Fig. 2). No
(1997) nomenclature) were still subject to large varia- increase in the cardiopulmonary toxicity and difference
tions of the contouring. This was related to the unclear in quality of life two years after treatment in comparison
definition of the upper border of those LNS. Recom- with non-irradiated patients were demonstrated in this
mended PORT CTV in this study included: bronchial study (Kepka et al. 2011). We probably need more data
stump, any possible extension to the mediastinal pleura on the safety of such reduced elective fields for PORT, as
adjacent to the tumour bed, any involved LNS; and well as more data on the possibility of further reduction
(regardless of involvement) ipsilateral hilum, subcarinal of this volume. In the meantime, it is probably reasonable
(LNS 7), lower paratracheal (4R and 4R) LNS. For left option for those who treat pN2 patients with PORT to use
side, subaortic (5) and paraaortic (6) LNS had to be the volumes as proposed above.
included. All LNS located between two non-contiguous For pN0 and pN1 patients, PORT is not used, as its
involved LNS had also to be included in the CTV deleterious effect on the survival has been demon-
(Spoelstra et al. 2010). Independently of this study, strated before. However, if one is using PORT, pos-
recently published prospective trial that evaluated pro- sibly in case of microscopically incomplete (R1)
spectively the quality of life and cardiopulmonary mor- resection, the results of randomized trial published by
bidity of pN2 patients receiving PORT in comparison Trodella et al. (2002) may be considered. In this
with pN1 patients in whom PORT was not given used study, only bronchial stump and ipsilateral hilar
very similar PORT volume definition. CTV included region were included in the CTV of pN0 completely
bronchial stump and LNS with presence of metastases in resected patients. The irradiated patients had 2% of
pathologic examination. Additionally, the uninvolved local failure compared to 23% at five years for control
LNS with the highest probability of microscopic group, and it was related to the border-line significant
involvement, namely ipsilateral hilum, subcarinal nodes 5-year survival improvement (p = 0.048).
196 L. Kepka and M. Kolodziejczyk

The use of PORT after incomplete resection (R1 or for set-up errors. We will briefly report on how to deal
R2) in subject to fewer controversies, as it is a com- with both of these issues in the target definition.
mon belief, that additional local treatment may pre-
vent or delay recurrence. It is beyond the scope of this
chapter to discuss the efficacy of PORT in such case 4.1 Internal Target Volume: Respiratory
scenario. However, we do not have clear and evi- Motion Management
dence-based guidelines for such indications. The
experts of the International Association for the Study The extent of lung tumor mobility varies individually and
of Lung Cancer (IASLC) (Rami-Porta et al. 2005) depends on many factors. Therefore, it should be esti-
provided a definition of incomplete resection after mated individually and done separately for both tumor
resection of lung cancer carried out with curative and lymph nodes (van Sornsen de Koste et al. 2002,
intent. It contains not only the cases of R1 resection 2003). The ‘‘population-based’’ margins should be
with involvement of resection margins (bronchial, replaced by the ‘‘patient-based’’ margins. Numerous
vascular, parenchymal, or parietal), but also a positive approaches have been used for determination of the
lymph node that was not removed, extracapsular extent of respiratory motion, including the fluoroscopy,
nodal extension, and positive pleural or pericardial slow CT scans, multiple planning scans, CT scans gen-
effusion. It is hardly conceivable to conduct and plan erated at maximal inspiration and expiration, and
any radiation volume for the last indication of dis- recently one approach that superseded all former meth-
semination of cancer to the visceral cavities. How- ods—respiratory-correlated four-dimensional (4D)-CT
ever, all other indications may be divided in cases of scanning approach (Slotman et al. 2006). The estimation
non-radical (R1) tumor and non-radical (R1) lymph of the tumor motion at the fluoroscopy is definitely an
node resections; and PORT represents the conceiv- outdated approach, as this method does not lead to the
able therapeutic option for both types. For R1 and N0 three-dimensional display of the extent and shape of the
disease, it is not formally recommended to use ENI, tumor displacement. Slow CT scanning with time of the
as its use is controversial. In such cases, the CTV acquisition of one scan of about 4 s (average time of
volume should include the area of the positive mar- respiratory cycle) allowed capturing a respiratory
gin. However, thoughtful clinical judgment should be movement of the tumor, but it led to a ‘‘blurred’’ image,
used when making a decision about a possible which was a big inconvenience for contouring. Approa-
inclusion of the nodal area in the CTV, taking into ches based on multiple scanning are currently replaced by
account, the predictable toxicity of such an approach, commercially available 4D-CT scanning. In this method,
quality of surgical staging, and imaging studies per- a respiratory signal is synchronously recorded during
formed. This caution is based on the findings that the image acquisition. The imaging data is retrospectively
proximal microscopic tumor extension related to the sorted to create a 4D dataset which contains 3D datasets
R1 resection on the bronchial wall is associated with for 10 phase bins within a respiratory cycle. It leads to the
an increased risk of lymph node invasion (Kara et al. construction of the internal target volume (ITV) that
2002). For extracapsular nodal extension, the irradi- encompasses all motion and shape changes over the
ation of mediastinum is a reasonable option according respiratory cycle. Such construction of the ITV may be a
to the rules described above with the dose boost (up to laborious process that requires a delineation of the GTV
60 Gy) done to the LNS with R1 findings. The rules on the 10 separate datasets for 10 captured bins of the
and controversies described in the paragraph on the respiratory cycle. There are several ways to overcome
contouring of particular LNS apply here. this major drawback of the 4D techniques which repre-
sents an increase in the workload at the radiation oncol-
ogy department. One solution is to only use of two
4 Planning Target Volume Definition extreme phases of the respiratory cycle, i.e., the end-
expiration and the end-inspiration (Rietzel et al. 2006).
Margins added to the CTV for creation of the plan- This method speeds up the work of radiation oncology
ning target volume (PTV) in NSCLC have two main staff, however; it may introduce errors such as underes-
components: (1) for motion (mainly respiratory, but timation of the changes of the shape of the tumor between
also cardiac beats) of tumor and organs at risk and (2) delineated phases (possible ignorance of the hysteresis
Target Volume Definition in Non-Small Cell Lung Cancer 197

effect) and introduction of artifacts by interpolation together with tumor/lymph nodes mobility and (at
(Persson et al. 2010). Another help in contouring on the lesser extent) anatomical changes during treatment.
4D-CT dataset is using the post-processing tool of max- To account for those uncertainities, the population-
imum intensity projection-MIP (the use of the maximum based recipes for margins between CTV and PTV were
density in Hounsfield units on all CT scans for automatic formulatd, from which the Van Herk et al. (2000) for-
generation of the target) for rapid computation of the mula has become the standard for margins definition
ITV. However, this method is far from being perfect and (Sonke and Beldernos 2010). In order to deliver at least
is only suitable for tumors surrounded by pulmonary 95% of dose to the CTV for 90% of patients the margin
parenchyma, and even in those cases, the careful visual should be calculated according to the formula:
inspection of the GTV on all datasets of respiratory cycle X p 2 
M ¼ 2:5 þb r þ r2p  brp
should be done (Rietzel et al. 2008). The ITV computed
from 4D-CT including all tumor motion leads to the P
where M is the CTV to PTV margin; the total standard
inclusion of the large volume of lung tissue in the PTV.
deviation (SD) of the systematic errors; r the total SD of
Thus—in case of the major tumor mobility—the gated
the random errors; rp the width of the penumbra mod-
radiotherapy techniques are in use, as, for example, the
eled by a cumulative Gaussian, and b is the value of the
irradiation at the end of expiration. In such scenario, GTV
inverse cumulative standard normal distribution at the
is delineated in the dataset corresponding to the needed
prescribed PTV minimum dose level (van Herk et al.
respiratory phase.
2000). It shows that systemtic error (that occurs at the
It was shown that the impact of the respiratory
planning stage) impacts much more than random errors
induced motion on the accumulated GTV dose is rel-
occurring at the treatment delivery. For lung cancer, the
atively small, providing that the time-averaged mean
one SD for systematic and random set-up error is up to
position of the GTV is correctly positioned. Wolthaus
4–5 mm (Hurkmans et al. 2001 and Borst et al. 2007);
et al. (2006) have developed a concept of mid-venti-
the one SD for systematic and random respiratory
lation scan, which is extracted from 4D-CT dataset and
motion displacement is up to 7 mm (Sonke and
represents an average position of the tumor during a
Beldernos 2010). For calculation of the margin, the all
respiratory cycle. On such chosen 3D CT frame, the
components of the inaccuracies should be considered.
GTV and subsequent CTV are delineated. The appro-
However, the set-up and motion errors are generally
priate margins for tumor motion and other uncertainties
uncorrelated, and a linear addition of these two com-
margins are calculated. Such constructed final PTV is
ponents is not correct (van Herk et al. 2000). Thus the
smaller than ITV computed from a whole the 4D-CT
different sources of geometrical uncertainities should be
dataset and it still adequately accounts for motion.
added in quadrature not in a linear fashion. It is common
Those considerations of the tumor mobility require
to add the margin directly to the CTV without separating
the use of complex equipment which, even if it is com-
two components (for ITV and set-up). In most clinical
mercially available, may be too expensive and/or too
scenarios of lung cancer, the margin for PTV does not
time consuming for use in all cases. When such equip-
exceed 1 cm. At the planning stage, the population-
ment is not available, at least an estimation of the tumor
based margins should be established. The repetitive
mobility for tumors located in the lower lobes and/or for
imaging during treatment, using multiple available
patients with restricted pulmonary reserves is needed.
technologies of image-guided radiotherapy, may lead to
The recognized mobility of lymph nodes of about
the individualization of the margins by individual
0.5 cm (for most) and about 1.0 cm for subcarinal nodes
quantification of the inter- and intrafractional errors
should be considered (van Sornsen de Koste et al. 2002).
which has now become a new emerging standard.

4.2 Margins for Set-up


References
Set-up error defined as a difference of bony anatomy
Belderbos J, Uitterhoeve L, van Zandwijk N et al (2007)
between plan and treatment represents an immanent Randomised trial of sequential versus concurrent chemo-
part of the geometrical uncertainty for any treatment radiotherapy in patients with inoperable non-small cell lung
with radiation. For lung cancer it should be considered cancer (EORTC 08972–22973). Eur J Cancer 43:114–121
198 L. Kepka and M. Kolodziejczyk

Belderbos JSA, Kepka L, Kong F-M, Martel MK, Videtic comparative analysis of toxicities and clinical outcomes.
GMM, Jeremic B (2008) Report from the international Radiother Oncol 95:178–184
atomic energy agency (IAEA) consultants’ meeting on Fournel P, Robinet G, Thomas P, Groupe Lyon-Saint-Etienne
elective nodal irradiation in lung cancer: non-small cell lung d’Oncologie Thoracique-Groupe Français de Pneumo-
cancer (NSCLC). Int J Radiat Oncol Biol Phys 72:335–342 Cancérologie (2005) Randomized phase III trial of
Black QC, Grills IS, Kestin LL (2004) Defining a radiotherapy sequential chemoradiotherapy compared with concurrent
target with positron emission tomography. Int J Radiat chemoradiotherapy in locally advanced non-small-cell lung
Oncol Biol Phys 60:1272–1282 cancer: Groupe Lyon-Saint-Etienne d’Oncologie Thoraci-
Boellard R, O’Doherty MJ, Weber WA et al (2010) FDG PET que-Groupe Français de Pneumo-Cancérologie NPC 95-01
and PET/CT: EANM procedure guidelines for tumour PET Study. J Clin Oncol 23:5910–5917
imaging: version1.0. Eur J Nucl Med Mol Imaging 37: Furuse K, Fukuoka M, Kawahara M et al (1999) Phase III study
181–200 of concurrent versus sequential thoracic radiotherapy in
Borst GR, Sonke JJ, Betgen A et al (2007) Kilo-voltage cone- combination with mitomycin, vindesine, and cisplatin in
beam computed tomography set-up measurements for lung unresectable stage III non–small-cell lung cancer. J Clin
cancer patients; first clinical results and comparison with Oncol 17:2692–2699
electronic portal imaging device. Int J Radiat Oncol Biol Giraud P, Antoine M, Larrouy A et al (2000) Evaluation of
Phys 68:555–561 microscopic tumor extension in non-small cell lung cancer
Bowden P, Fisher R, MacManus M et al (2002) Measurement for three-dimensional conformal radiotherapy treatment
of lung tumor volumes using three-dimensional computer planning. Int J Radiat Oncol Biol Phys 48:1015–1024
planning software. Int J Radiat Oncol Biol Phys 53:566–573 Giraud P, Elles S, Helfre S et al (2002) Conformal radiotherapy
Bradley J, Bae K, Choi N et al (2010) A phase II comparative for lung cancer: different delineation of the gross tumor
study of gross tumor volume definition with or without PET/ volume (GTV) by radiologists and radiation oncologists.
CT fusion in dosimetric planning for non-small cell lung Radiother Oncol 62:27–36
cancer (NSCLC): primary analysis of radiation therapy Giraud P, De Rycke Y, Lavole A et al (2006) Probability of
oncology group (RTOG) 0515. Int J Radiat Oncol Biol Phys mediastinal involvement in non-small-cell lung cancer: a
(in press) statistical definition of the clinical target volume for
Chapet O, Kong F-M, Quint LE et al (2005) CT-based 3-dimensional conformal radiotherapy? Int J Radiat Oncol
definition of thoracic lymph node stations: an Atlas from Biol Phys 64:127–135
the University of Michigan. Int J Radiat Oncol Biol Phys Greco C, Rosenzweig K, Cascini GL, Tamburini O (2007)
63:170–178 Current status of PET/CT for tumour volume definition in
Curran WJ, Scott CB, Langer CJ et al (2003) Long-term benefit radiotherapy treatment planning for non-small cell lung
is observed in a phase III comparison of sequential vs cancer (NSCLC). Lung Cancer 57:125–134
concurrent chemoradiation for patients with unresected Grills IS, Yan D, Martinez AA et al (2003) Potential for
stage III NSCLC: RTOG 9410. Proc Am Soc Clin Oncol reduced toxicity and dose escalation in the treatment of
22:621 (abstr 2499) inoperable non-small-cell lung cancer: a comparison of
de Langen AJ, Raijmakers P, Riphagen I, Paul MA, Hoekstra intensity-modulated radiation therapy (IMRT), 3D confor-
OS (2006) The size of mediastinal lymph nodes and its mal radiation, and elective nodal irradiation. Int J Radiat
relation with metastatic involvement: a meta-analysis. Eur J Oncol Biol Phys 57:875–890
Cardiothorac Surg 29:26–29 Grills IS, Fitch DL, Goldstein NS et al (2007) Clinicopatho-
de Ruysscher D, Wanders R, van Haren E et al (2008) HI- logic analysis of microscopic extension in lung adenocar-
CHART: a phase I/II study on the feasibility of high dose cinoma: defining clinical target volume for radiotherapy. Int
continuous hyperfractionated accelerated radiotherapy in J Radiat Oncol Biol Phys 69:334–341
patients with inoperable non-small cell lung cancer. Int J Huber RM, Flentje M, Schmidt M, Bronchial Carcinoma
Radiat Oncol Biol Phys 71:132–138 Therapy Group et al (2006) Simultaneous chemoradiother-
de Ruysscher D, Faivre-Finn C, Nestle U et al (2010) European apy compared with radiotherapy alone after induction
organization for research and treatment of cancer recom- chemotherapy in inoperable stage IIIA or IIIB non-small-
mendations for planning and delivery of high dose, high- cell lung cancer: study CTRT99/97 by the bronchial
precision of radiotherapy for lung cancer. J Clin Oncol carcinoma therapy group. J Clin Oncol 24(27):4397–4404
28(36):5301–5310 Hurkmans CW, Remeijer P, Lebesque JV, Mijnher BJ (2001)
Devic S, Tomic N, Faria S et al (2010) Defining radiotherapy Set-up verification using portal imaging; review of current
target volumes using 18F-fluoro-deoxy-glucose positron clinical practice. Radiother Oncol 58:105–120
emission tomography/computed tomography: still a International Commission on Radiation Units and Measure-
Pandora’s box? Int J Radiat Oncol Biol Phys 78:1555–1562 ments (1993) ICRU Report 50. Prescribing, recording and
Dillman RO, Herndon J, Seagren SL, Eaton WL Jr, Green MR reporting photon beam therapy. ICRU Bethesda, MD, USA
(1996) Improved survival in stage III non–small-cell lung International Commission on Radiation Units and Measure-
cancer: seven-year follow-up of cancer and leukemia group ments (1999) ICRU Report 62. Prescribing, recording and
B (CALGB) 8433 trial. J Natl Cancer Inst 88:1210–1215 reporting photon beam therapy. ICRU Bethesda, MD, USA
Fernandes AT, Shen J, Finlay J et al (2010) Elective nodal Kara M, Dizbay Sak S, Orhan D, Yavuzer S (2000) Changing
irradiation (ENI) vs. involved field radiotherapy (IFRT) for patterns of lung cancer;(3/4 in.) 1.9 cm; still a safe length
locally advanced non-small cell lung cancer (NSCLC): a for bronchial resection margin? Lung Cancer 30:161–168
Target Volume Definition in Non-Small Cell Lung Cancer 199

Kara M, Dizbay Sak S, Orhan D, Kavukcu S (2001) Proximal Persson GF, Nygaard DE, Brink C et al (2010) Deviations in
bronchial extension with special reference to tumor delineated GTV caused by artefacts in 4DCT. Radiother
localization in non-small cell lung cancer. Eur J Cardi- Oncol 96:61–66
othorac Surg 20:350–355 PORT Meta-analysis Trialists Group (1998) Postoperative
Kara M, Dikmen E, Kilic D et al (2002) Prognostic implication radiotherapy in non-small-cell lung cancer: systematic
of microscopic proximal bronchial extension in non-small review and meta-analysis of individual patient data from
cell lung cancer. Ann Thorac Surg 74:348–354 nine randomized controlled trials. Lancet 352:257–263
Kelsey CR, Marks LB, Glatstein E (2009) Elective nodal Rami-Porta R, Wittekind C, Goldstraw P, International
irradiation for locally advanced non-small-cell lung cancer: Association for the Study of Lung Cancer (IASLC) Staging
it’s called cancer for a reason. Int J Radiat Oncol Biol Phys Committee (2005) Complete resection in lung cancer
73:1291–1292 surgery: proposed definition. Lung Cancer 49:25–33
Kepka L, Bujko K, Garmol D et al (2007) Delineation variation Rietzel E, Liu AK, Doppke KP et al (2006) Design of 4D
of lymph node stations for treatment planning in lung cancer treatment planning target volumes. Int J Radiat Oncol Biol
radiotherapy. Radiother Oncol 85:450–455 Phys 66:287–295
Kepka L, Bujko K, Zolciak-Siwinska A (2008) Risk of isolated Rietzel E, Liu AK, Chen GTY, Choi N (2008) Maximum
nodal failure for non-small cell lung cancer (NSCLC) treated intensity volumes for fast contouring of lung tumors
with the elective nodal irradiation (ENI) using 3D-conformal including respiratory motion in 4DCT planning. Int J Radiat
radiotherapy (3D-CRT) techniques. Acta Oncol 47:95–103 Oncol Biol Phys 71:1245–1252
Kepka L, Tatro D, Moran JM et al (2009a) Designing targets Rocmans P, Emami B, Cox JD et al (1991) Quality control in
for elective nodal irradiation in lung cancer radiotherapy: a NSCLC treatment: a consensus report. Lung Cancer 7:
planning study. Int J Radiat Oncol Biol Phys 73:1397–1403 S19–S20
Kepka L, Tyc-Szczepaniak D, Bujko K (2009b) Dose per Rowell NP, Williams CJ et al (2001) Radical radiotherapy for
fraction escalation of accelerated hypofractionated three- stage I/II non-small cell lung cancer in patients not
dimensional conformal radiotherapy in locally advanced sufficiently fit or declining surgery (medically inoperable).
non-small cell lung cancer. J Thorac Oncol 4:853–861 Cochrane Database Syst Rev 2:CD002935
Kepka L, Bujko K, Orlowski T et al (2011) Cardiopulmonary Rusch VW, Asamura H, Watanabe H et al Members of IASLC
morbidity and quality of life in non-small cell lung cancer Staging Committee (2009) . The IASLC lung cancer staging
patients treated with or without postoperative radiotherapy. project: a proposal for a new international lymph node map
Radiother Oncol 98:238–243 in the forthcoming seventh edition of the TNM classifica-
Kołodziejczyk M, Kepka L, Dziuk M et al (2010) Impact of tion for lung cancer. J Thorac Oncol 4:568–577
[18F]fluorodeoxyglucose PET-CT staging on treatment Saito M, Yokoyama A, Kurita Y et al (2000) Treatment of
planning in radiotherapy incorporating elective nodal irra- roentgenographically occult endobronchial carcinoma with
diation for non-small cell lung cancer: a prospective study. external beam radiotherapy and intraluminal low-dose rate
Int J Radiat Oncol Biol Phys (in press) brachytherapy: a second report. Int J Radiat Oncol Biol
Lagerwaard FJ, van de Vaart PJ, Voet PW et al (2002) Can Phys 47:673–680
errors in reconstructing pre-chemotherapy target volumes Sause W, Kolesar P, Taylor SI et al (2000) Final results of
contribute to the inferiority of sequential chemoradiation in phase III trial in regionally advanced unresectable non–
stage III non-small cell lung cancer (NSCLC)? Lung Cancer small-cell lung cancer: radiation therapy oncology group,
38:297–301 eastern cooperative oncology group, and southwest oncol-
Le Chevalier T, Arriagada R, Quoix E et al (1991) Radiother- ogy group. Chest 117:358–364
apy alone versus combined chemotherapy and radiotherapy Silvestri GA, Gould MK, Margolis ML et al (2007) Noninvasive
in nonresectable non–small-cell lung cancer: first analysis staging of non-small cell lung cancer: ACCP evidence-based
of a randomized trial in 353 patients. J Natl Cancer Inst clinical practice guidelines (2nd edition). Chest 132:178–201
83:417–423 Slotman BJ, Lagerwaard FJ, Senan S (2006) 4D imaging for
Macmanus M, Nestle U, Rosenzweig KE et al (2009) Use of target definition in stereotactic radiotherapy for lung cancer.
PET and PET-CT for radiation therapy planning: IAEA Acta Oncol 45:966–972
expert report 2006–2007. Radiother Oncol 91:85–94 Sonke JJ, Beldernos J (2010) Adaptive radiotherapy for lung
Mountain CF, Dresler CM (1997) Regional lymph node cancer. Sem Radiat Oncol 20:94–106
classification for lung cancer staging. Chest 111:1718–1723 Sornsen van, de Koste JR, Lagerwaard FJ, Nijssen-Visser MRJ
Naruke T, Suemasu K, Ishikawa S (1978) Lymph node et al (2002) What margins are necessary for incorporating
mapping and curability at various levels of metastasis in mediastinal nodal mobility into involved-field radiotherapy
resected lung cancer. J Thorac Cardiovasc Surg 76:833–839 for lung cancer? Int J Radiat Oncol Biol Phys 53:1211–1215
Nestle U, Kremp S, Grosu A (2006) Practical integration of Sornsen van, de Koste JR, Lagerwaard FJ, Nijssen-Visser MRJ
[18F]-FDG-PET and PET-CT in the planning of radio- et al (2003) Tumor location cannot predict the mobility of
therapy for non-small cell lung cancer (NSCLC): the lung tumors: a 3D analysis of data generated from multiple
technical basis, ICRU-target volumes, problems, perspec- CT scans. Int J Radiat Oncol Biol Phys 56:348–354
tives. Radiother Oncol 81:209–225 Spoelstra FOB, Senan S, Le Pechoux C et al (2010) Variations
Pasic A, Brokx HA, Comans EF (2005) Detection and staging in target volume definition for postoperative radiotherapy in
of preinvasive lesions and occult lung cancer in the central stage III non-small cell lung cancer: analysis of an
airways with 18F-fluorodeoxyglucose positron emission international contouring study. Int J Radiat Oncol Biol
tomography: a pilot study. Clin Cancer Res 11:6186–6189 Phys 76:1106–1113
200 L. Kepka and M. Kolodziejczyk

Steenbakkers RJ, Duppen JC, Fitton I et al (2005) Observer Cancer Group et al (2007) Randomized controlled trial of
variation in target volume delineation of lung cancer related resection versus radiotherapy after induction chemotherapy
to radiation oncologist–computer interaction: a ‘‘Big in stage IIIA-N2 non-small-cell lung cancer. J Natl Cancer
Brother’’ evaluation. Radiother Oncol 77:182–190 Inst 99:442–450
Steenbakkers RJ, Duppen JC, Fitton I et al (2006) Reduction of Videtic GM, Rice TW, Murthy S et al (2008) Utility of positron
observer variation using matched CT-PET for lung cancer emission tomography compared with mediastinoscopy for
delineation: a three-dimensional analysis. Int J Radiat Oncol delineating involved lymph nodes in stage III lung cancer:
Biol Phys 64:435–438 insights for radiotherapy planning from a surgical cohort.
Stroom J, Blaauwqeers H, van Baardwijk A et al (2007) Int J Radiat Oncol Biol Phys 72:702–706
Feasibility of pathology-correlated lung imaging for accu- Vokes EE, Herndon JE 2nd, Kelley MJ, Cancer, Leukemia
rate target definition of lung tumors. Int J Radiat Oncol Biol Group B et al (2007) Induction chemotherapy followed by
Phys 69:267–275 chemoradiotherapy compared with chemoradiotherapy
Tai P, Yu E, Battista J, van Dyk J (2004) Radiation treatment of alone for regionally advanced unresectable stage III Non-
lung cancer–patterns of practice in Canada. Radiother Oncol small-cell lung cancer: cancer and leukemia group B. J Clin
71:167–174 Oncol 25:1698–1704
Thomas M, Rübe C, Hoffknecht P, German Lung Cancer Wolthaus JWH, Schneider C, Sonke JJ et al (2006) Mid-
Cooperative Group et al (2008) Effect of preoperative ventilation CT scan construction from four dimensional
chemoradiation in addition to preoperative chemotherapy: a respiration-correlated CT scans for radiotherapy planning of
randomised trial in stage III non-small-cell lung cancer. lung cancer patients. Int J Radiat Oncol Biol Phys
Lancet Oncol 9:636–648 65:1560–1571
Trodella L, Granone P, Valente S et al (2002) Adjuvant Wu K, Ung YC, Hornby J et al (2010) PET CT thresholds for
radiotherapy in non-small cell lung cancer with pathological radiotherapy target definition in non-small cell lung cancer:
stage I: definitive results of a phase III randomized trial. how close are we to the pathologic findings? Int J Radiat
Radiother Oncol 62:11–19 Oncol Biol Phys 77:699–706
van de Steene J, Linthout N, De Mey J et al (2002) Definition of Yuan S, Meng X, Yu J et al (2007a) Determining optimal
gross tumor in lung cancer: inter-observer variability. clinical target volume margins on the basis of microscopic
Radiother Oncol 62:37–49 extracapsular extension of metastatic nodes in patients with
van Herk M, Remeijer P, Rasch C, Lebesque JV (2000) The non-small-cell lung cancer. Int J Radiat Oncol Biol Phys
probability of correct target dosage: dose population 67:727–734
histograms for deriving treatment margins in radiotherapy. Yuan S, Sun X, Li M et al (2007b) A randomized study of
Int J Radiat Oncol Biol Phys 47:1121–1135 involved-field irradiation versus elective nodal irradiation in
van Loon J, Siedschlag C, Stroom J et al (2010) Microscopic combination with concurrent chemotherapy for inoperable
disease extension in three dimensions for non-small cell lung stage III non-small cell lung cancer. Am J Clin Oncol
cancer: development of a prediction model using pathology 30:239–244
validated positron emission tomography and computed Zatloukal P, Petruzelka L, Zemanova M et al (2004) Concur-
tomography features. Int J Radiat Oncol Biol Phys (in press) rent versus sequential chemoradiotherapy with cisplatin and
van Meerbeeck JP, Kramer GW, van Shil PE, European vinorelbine in locally advanced non-small-cell lung cancer:
Organisation for Research, Treatment of Cancer-Lung A randomized study. Lung Cancer 46:87–98
The Radiation Target in Small-Cell
Lung Cancer
Gregory M. M. Videtic

Contents Abstract
What constitutes the appropriate target in the
1 Introduction to the Role of Radiotherapy treatment of small-cell lung cancer is an area of
in SCLC .................................................................... 201 active investigation. Advances in the last 30 years
2 Historic Trends in Lung Cancer in radiologic and nuclear imaging as well as in
Target Definition...................................................... 202 radiotherapy delivery have prompted revision of
3 SCLC Target Definition and the Impact the classically defined target which was designed
of CHT ...................................................................... 203 to be ‘‘tolerable’’ and encompass visible tumor
4 SCLC Target Definition and ENI.......................... 206 and potential areas of microscopic disease. Cur-
rent trends are to minimize normal tissue irradi-
5 SCLC Target Definition and FDG-PET ............... 208
ation and more precisely define the extent of
6 SCLC Target Definition in Extensive Disease ..... 208 potential microscopic spread in order to optimize
7 SCLC Target Definition in Current Trials .......... 209 target volume. This chapter will review historic
8 Conclusions ............................................................... 209 trials and ongoing studies to provide a compre-
hensive understanding on the evolution of the
References.......................................................................... 209
radiotherapy target in small-cell lung cancer.

1 Introduction to the Role


of Radiotherapy in SCLC

Thoracic radiotherapy (TRT) is an integral com-


ponent in the standard management of patients
presenting with small-cell lung cancer (SCLC). Its
role in improving survival and local control in the
treatment of limited stage disease (LS-SCLC) has
been confirmed in randomized clinical trials over
the past 25 years and in subsequent meta-analyses
(Cooper and Spiro 2006; Lee et al. 2006; Faivre-
Finn et al. 2005a, b; Socinski and Bogart 2007;
G. M. M. Videtic (&) Curran 2001; de Ruysscher and Vansteenkiste
Department of Radiation Oncology/T28, 2000). These reports have shown that the addition
Taussig Cancer Institute, Cleveland Clinic Foundation,
of TRT to combination chemotherapy (CHT) sig-
9500 Euclid Avenue, Cleveland, OH 44195, USA
e-mail: videtig@ccf.org nificantly reduces the risk of loco-regional failure

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 201
DOI: 10.1007/174_2011_251,  Springer-Verlag Berlin Heidelberg 2011
202 G. M. M. Videtic

(Bleehen et al. 1983; Bunn et al. 1987; Mira et al.


1982; Perez et al. 1984; Perry et al. 1987), and two 2 Historic Trends in Lung Cancer
meta-analyses have shown an absolute long-term Target Definition
survival gain of 5% (Pignon et al. 1992; Warde
and Payne 1992). The role of TRT in extensive In principle, when planning curative RT for lung
stage disease (ES-SCLC) is well established as an cancer (whether NSCLC or SCLC) the target volume
important palliative modality but less so for of tissue irradiated to a high-dose should only
potentially influencing overall survival. In that encompass the entire tumor and any microscopic
respect, a provocative phase III study published in extension of disease, and be kept as small as possible
1997 showed that the addition of TRT improved to minimize damage to normal tissues. From 1960s to
survival in patients selected by response to che- 1990s, lung cancer volumes typically encompassed
motherapy (Jeremic et al. 1999). Prophylactic the definable lung tumor and any overtly involved
cranial irradiation (PCI) has a well-defined role in lymph nodes (LNs) as discernable from chest radio-
LS-SCLC, with evidence showing a *50% graphs and also included all the regional nodes con-
reduction in brain metastasis rates and an absolute sidered at risk, which were generally those in the
improvement in overall survival of approximately mediastinum and the bilateral hilar and supraclavic-
5% (Auperin et al. 1999). More recently, favorable ular (SCF) regions. This approach assumed that all the
results for PCI with respect to survival and pre- regional LNs, even if seeming clinically uninvolved,
vention of symptomatic brain disease were should be irradiated in order to treat potential
observed in a phase III study of ES-SCLC patients microscopic spread since imaging-based definition of
receiving this therapy after demonstrating any disease extent was crude. This target often involved a
response to chemotherapy (Slotman et al. 2007). fairly large volume of the chest being irradiated,
How to best deliver potentially curative radio- which was usually done with relatively static, ‘‘for-
therapy (RT) remains an active area of investigation mulaic’’ field arrangements such as opposed anterior–
and debate. For example, in the setting of LS-SCLC, posterior fields, followed by a boost to involve tumor
there is evidence to support early versus later initia- and nodes using oblique fields sparing the spinal cord
tion of TRT with concurrent CHT because of a (Videtic et al. 2008).
favorable impact on survival, but this is not without The rapid evolution of radiologic imaging over the
controversy (Samson et al. 2007; Fried et al. 2004; past 20–30 years has allowed clinicians to move away
Huncharek and McGarry 2004; Pijls-Johannesma from the limitations of the chest radiograph to highly
et al. 2007; Spiro et al. 2006; Jeremic 2006). The sophisticated means of tumor definition, whether by
optimal dose of TRT to deliver also remains strongly computed tomography (CT) scans of the chest or
debated and is currently the subject of two ongoing more recently, by positron emission tomography
randomized studies, but the most current standard is (PET) scan. This had triggered a parallel shift in
based on a pivotal phase III study that demonstrated radiation practice because these technological
survival benefits using 45 Gy over a 3-week course of advances have permitted improved and potentially
hyperfractionated (twice-daily) TRT (Turrisi et al. more accurate definition of the lung and LNs clini-
1999). Lastly, what exactly constitutes the appropriate cally involved in tumor. Such refinements in RT
target volume to be irradiated in LS-SCLC within a planning have been matched by standardization of
given radiochemotherapy program also has not been dose reporting criteria, target definitions, and normal
clearly defined. There is no doubt that questions on structure labeling by keeping with updates from the
target definition are dependent in some fashion on International Commission on Radiation Units and
other components of RT for SCLC, such as total dose, Measurements (International Commission on Radia-
fractionation, and timing of administration with tion Units and Measurements 1993; International
respect to CHT. That said, the purpose of this chapter Commission on Radiation Units and Measurements
is to summarize perspectives on target delineation in 1999). Terms such as gross tumor volume (GTV)
TRT for SCLC and make reference to prospective and indicating detectable or visible disease; clinical target
retrospective reports that may inform current practice volume (CTV), containing the GTV with sufficient
in the management of SCLC. margins to account for subclinical disease extension;
The Radiation Target in Small-Cell Lung Cancer 203

and planning target volume (PTV), a geometrical involving parenchyma and MLNs, and surrounding
parameter obtained by adding adequate margins normal structures. This explains in part why the
around the CTV to account for uncertainties linked to specific question of nodal irradiation was often not a
set-up errors and organ motion, are now routinely question to be separated out from defining treatment
assigned to structures during RT target delineation. to the primary disease (Videtic et al. 2008).
Even more recently, accounting for organ-motion- With these historical trends in mind, it will be
associated geometric uncertainties and daily set-up noted that this chapter reflects results from fact clin-
errors have become more sophisticated. An internal ically oriented questions with respect to target defi-
target volume (ITV) reflecting potential tumor dis- nition since they have been the common ones
placements in space is now regularly defined to addressed in prospective studies of TRT. As refer-
minimize PTV expansions for motion. Refinements in ence, Table 1 provides a comprehensive list of com-
the means of verification imaging before and during pleted or ongoing clinical trials over the last 30 years
RT delivery are now being accomplished by sophis- and their respective definitions of the target. Not-
ticated means of image-guided RT (IGRT) systems. withstanding the range of technological innovations
IGRT may permit real time adjustment of RT as in imaging and RT delivery and their rapid imple-
needed on the basis of and according to therapy- mentation with respect to modifying target definition,
induced tumor changes (Aristei et al. 2010). there are very few studies reported on in this chapter
On the clinical front, evolving concepts in target showing the impact that technology has had on SCLC
definition for lung cancer have been especially TRT. To date, evidence is lacking for survival bene-
focused on the issue of mediastinal (regional) lymph fits by adopting these advances, although for many
node (MLN) irradiation. As noted above, radiation authors their impact on normal tissues has been as
oncologists historically defined the lung cancer portal critical as on tumor outcomes.
so that there was comprehensive inclusion of all MLN
stations irrespective of disease status [‘‘elective nodal
irradiation’’ (ENI)] (Emami 1996). However, it is now 3 SCLC Target Definition
becoming common to see omission of ENI in con- and the Impact of CHT
temporary clinical trials and routine treatment (Senan
and de Ruysscher 2005), despite the fact that there are The earliest questions relative to defining the optimal
only very limited prospective data outcomes to sup- treatment volume for SCLC were related to the use of
port this practice. A recently completed survey of 800 induction CHT prior to the start of TRT. Since sig-
radiation oncologists reveals in that regard in that 31, nificant tumor shrinkage can often occur with a
51, and 18% of respondents chose extensive, selective number of CHT cycles, this prompted the question of
or no ENI for SCLC, respectively (Kong et al. 2007). what should be designated as the appropriate TRT
The rationale underpinning omission of ENI has been volume to be treated: the pre- or the post-CHT vol-
the need to improve on the recognized high relapse ume. It is interesting to note that it is in this setting
rates at the site of the initial tumor after conventional that the only randomized clinical trial to date
TRT. Thus, the required dose escalation to potentially addressing the specific question of TRT treatment
improve local control has been judged to be feasible volume in SCLC has been conducted. This study,
only if clinicians were to treat gross disease only, i.e., performed by the Southwest Oncology Group
the primary and any involved nodes, termed an (SWOG) and published in 1987 (Kies et al. 1987),
involved-field, or non-ENI, approach. The expecta- involved 466 patients and had a complex randomi-
tion is that by reducing the treated volume, one could zation schema based on response; in short, patients
reduce the toxicity to the critical normal structures with a partial response or stable disease after four
and therefore allow delivery of higher TRT doses. cycles of CHT (non-platinum based) were random-
With these clinical goals in mind, at the time of SCLC ized to RT fields based either on the pre- or the post-
diagnosis the distinction between primary tumor and CHT volume of disease. No statistical differences in
mediastinal disease is nonetheless sometimes difficult survival or recurrence patterns were noted as a func-
to establish, because the disease can appear as a tion of volume treated: for complete responders, with
conglomerate, relatively indistinct central tumor mass local recurrence rates of 50% with RT, and 72%
Table 1 Target volume definitions from completed and ongoing prospective studies in limited stage SCLC
204

Publication Author (ref.) Study type Planning Pre- or post- Overall target definition
year (2D, 3D) CHT primary
target
Ongoing CALGB30610/ Phase III 3D With cycle GTV by CT, and PET if available
RTOG0538 1 or 2 ITV for motion
CTV1-GTV ? ipsilateral hilum; CTV2-revised; GTV after CHT; otherwise no ENI
PTV 1 and 2- non-ITV or ITV based margins, between 15 and 5 mm, respectively
Ongoing EORTC- Phase III 3D With cycle 2 GTV by CT, and PET if available
CONVERT CTV = GTV ? 5 mm
PTV = 10 mm sup/inf; 8 mm lat
No ENI
2010 van Loon et al. Phase II 3D Post-CHT GTV and PTV defined by PET and CT
(2010) (median of If induction CHT, post-CHT volume considered GTV of primary tumor but for
18 days) MLNs, pre-CHT used
If PET negative in mdtsnm and CT positive, mdstnm not included in GTV.
Margin from GTV to CTV = 5 mm, from CTV to PTV = 5 mm for MLNs and
10 mm for primary
No. ENI
2007 Schild et al. Phase II 2D Post-CHT cycle Split course: cycle 4-primary tumor with ipsilateral hilar, mediastinal, and SCF
(2007) 4/5 of 6 nodes; cycle 5-cone-down to ‘‘reduced’’ mdstnm and only involved SCF
2006 Spiro et al. Phase III 2D Pre-CHT Arms 1, 2: primary
(2006) Tumor with margin ? entire mdstnm; SCF if involved
2006 Baas et al. Phase II 3D After cycle 1 Primary tumor ? all clinical and radiological involved lymph nodes with a short-
(2006) CHT axis diameter of [1 cm
2006 de Ruysscher Phase II 3D CT during the GTV = primary tumor and MLNs with a short-axis diameter of [1 cm
et al. (2006) first cycle of PTV = GTV ? margin
CHT
2004 Bogart et al. Phase II 2D or 3D After cycle 2 GTV = post induction lung tumor and involved MLN (pre- AND post-CHT)
(2004) CHT CTV1 [to 44 Gy] = GTV ? ipsilateral non-involved MLNs ? margin
CTV2 [to 70 Gy] = GTV ? ispilateral hilum
(continued)
G. M. M. Videtic
Table 1 (continued)
Publication Author (ref.) Study type Planning Pre- or post- Overall target definition
year (2D, 3D) CHT primary
target
2002 Takada et al. Phase III Not specified Pre-CHT Arm 1: RT with CHT cycle 1-primary disease with margin, ipsilateral hilum, entire
(2002) mdstnm, SCF if involved
Arm 2: RT after 4 cycles CHT-‘‘pretreatment tumor volume’’
2001 Skarlos et al. Randomized 2D Pre-CHT Arm 1: to 30 Gy-primary tumor ? entire mdtsnm ? bilateral hila; SCF if involved
(2001) phase II To 45 Gy-primary tumor
Arm 2: ‘‘initial tumor volume’’
1999 Turrisi et al. Phase III 2D Pre-CHT Primary tumor, bilateral mediastinal and ipsilateral hilar lymph nodes, SCF only if
(1999) involved, specifically: ‘‘inferior border … 5 cm below the carina or to a level
including ipsilateral hilar structures, whichever was lower’’.
1997 Jeremic et al. Phase III 2D Pre-CHT Arm 1: primary tumor, ipsilateral hilum, entire mdstnm, SCF only if involved
The Radiation Target in Small-Cell Lung Cancer

(1997) Arm 2: initial tumor volume


1997 Work et al. Phase III 2D (rare 3D) Pre-CHT Primary tumor, ipsilateral hilum, entire mdstnm, SCF only if involved
(1997)
1993 Murray et al. Phase III 2D (‘‘CT- Pre-CHT Arms 1, 2: Primary tumor with margin ? entire mdstm, SCF if involved
(1993) planning not
mandatory’’)
1987 Perry et al. Phase III 2D Pre-CHT Arms 1, 2: to 40 Gy-primary tumor with margin ? entire mdstnm, bilateral hila,
(1987) bilateral SCF
Boost to 50 Gy-residual disease on a mid-course CXR
1987 Kies et al. Phase III 2D Arm ‘‘wide- Primary tumor, ‘‘abnormal appearing lung’’, mdstnm, ‘‘low’’ SCF
(1987) field’’: pre-CHT
Arm ‘‘reduced
field’’: post-
CHT
1981 Perez et al. Phase III 2D Pre-CHT Arms 1, 2: primary tumor with margin ? entire mdstnm, bilateral hila, bilateral
(1981) SCF
CT computed tomography, PTV planning target volume, GTV gross tumor volume, CTV clinical target volume, ITV internal target volume, CHT chemotherapy, MLN mediastinal
lymph node, RT radiotherapy; SCF supraclavicular fossa, mdstnm mediastinum
205
206 G. M. M. Videtic

without RT, and for partial responders or those with This includes a series of studies looking at the impact
stable disease, local recurrences were 32% for pre- clinical trial violations on relapse patterns. Impaired
CHT volumes versus 28% for post-CHT volumes. local control and survival were found in trial patients
Thus post-CHT volumes were judged reasonable for in whom major RT protocol violations occurred
target delineation. The details of the portals used in (‘‘inadequate treatment portals’’), as reported both by
this protocol are relevant. As stated by the authors, the Perez et al. (1981) and by White et al. (1982) in their
volume as determined from a chest X-ray included: analyses of outcomes for SCLC patients enrolled on
the primary tumor, the surrounding abnormal lung, clinical trials in the 1970s. In contrast, in trials from
the low supraclavicular area, ‘‘which gave a very the 1980s when more sophisticated simulation tech-
large portal in some patients’’ (Kies et al. 1987). In niques became commonplace, especially CT-based
the pre-CHT arm, the X-ray was taken before the planning, Arriagada et al. (1991) and Brodin et al.
induction chemotherapy; in the reduced-field arm, the (1990) each found that tumor recurrences predomi-
post-induction X-ray served for planning. nated within the radiation port (i.e., the post-CHT
Several retrospective analyses have assessed pre- volume), suggesting that the dose delivered was more
versus post-CHT tumor volumes in the setting of TRT critical than target delineated.
for SCLC. Mira and Livingston (Mira and Livingston In summary, the only Phase III study ever con-
1980) assessed 17 LS-SCLC patients treated with ducted in target definition for LS-SCLC, a (partially)
post-CHT volumes and found that the majority who randomized trial by SWOG from the pre-platinum
failed in the chest also failed at the lung periphery CHT era, suggests that, in SCLC, the radiation target
(intrathoracic but outside the field) or with a malig- can be limited sufficiently to the residual gross tumor
nant pleural effusion but not nodal structures, sug- as defined clinically after administration of CHT.
gesting that pre-CHT volumes would provide Retrospective studies, on the other hand, have shown
improved local control rates. In contrast, an influential mixed results on the selection of the appropriate
study by Liengswangwong et al. at Mayo Clinic treatment volume for TRT. There is, however, a trend
supported the use of post-CHT volumes supporting target volumes limited to clinically defined
(Liengswangwong et al. 1994). Of 59 patients studied, disease only post-CHT.
28 were treated with field sizes that encompassed
post-CHT tumor volumes and 31 with fields that
encompassed pre-CHT tumor volumes (defined as a 4 SCLC Target Definition and ENI
volume that included at least a 1.5 cm margin on the
pre-CHT tumor volume). Nineteen patients had In as much as post-CHT pattern-of-failure data have
intrathoracic recurrence of disease as in-field failures suggested that the TRT target could be limited to the
stratified as follows: 10 of 31 patients treated with RT clinically definable disease (without specifically
fields that encompassed pre-CHT tumor volumes and addressing the ENI question), the trends in LS-SCLC
9 of 28 patients treated with RT fields that encom- management were increasingly to: (1) initiate TRT
passed post-CHT tumor volumes, suggesting no dif- with the earliest, if not the first cycle of CHT, using
ference between choice of volume for TRT. therefore a pre-CHT volume, and (2) work toward RT
Tada et al. looked at the patterns of recurrence in dose escalation. These goals were reflected in how
117 patients treated between 1986 and 1993 (Tada trials decided on the amount of MLN inclusion in the
et al. 1998). Patients were treated with a range of RT target as opposed to changing the primary tumor’s
doses and all with systemic CHT. There appeared to volume. As seen in Table 1, published studies as early
be more regional relapses in the upper mediastinum as 1981 started to show modest reductions in nodal
and supraclavicular fossae in those patients with sites included in the target volume by changing the
clinical N2 and N3 disease, prompting the authors to extent of LN inclusion. In the absence of supportive
recommend extended upper borders for N2 or N3 clinical data, such changes were effectively empirical
disease for unsuspected microscopic disease. Other as TRT dose escalation was being attempted.
means of understanding the impact that target defi- Study of Perez et al. (1981) and the randomized
nition may have on TRT fields have included retro- Cancer and Leukemia Group B (CALGB) study
spective studies looking at patterns of failure. published by Perry et al. (1987), volumes included the
The Radiation Target in Small-Cell Lung Cancer 207

gross tumor, ipsilateral hilum, bilateral mediastinal date directly addressing the issue of ENI in SCLC was
nodal chains, and both supraclavicular fossae (SCF). a phase II trial published by de Ruysscher et al.
A shift away from elective SCF inclusion was seen in (2006). The authors explicitly wished to evaluate the
Murray et al. (1993) Phase III LS-SCLC trial, where patterns of recurrence when ENI was omitted in
the target allowed only involved SCF, as did the patients with LS-SCLC. Twenty-seven patients
Aarhus trial by Work et al. (1997), in which the target received TRT with 45 Gy/30 fractions (1.5 Gy twice-
was defined by the pretherapy radiograph. Jeremic daily) concurrent with carboplatin and etoposide
et al. (1997) described therapy by anteroposterior- (CbE). Only the primary tumor and the positive nodal
posteroanterior fields to the gross tumor, ipsilateral areas on the pretreatment CT scan were irradiated.
hilum, entire mediastinum, and involved SCF. In the A PET scan was not performed. After a median time
landmark 1999 publication from Turrisi et al. (1999), of 18 months post-RT, 7 patients developed a local
the target which was defined as gross tumor by CT recurrence. Three patients (11%) developed an iso-
scanning, included full mediastinum and ipsilateral lated nodal failure, all of them in the ipsilateral SCF.
hilum, but no elective irradiation of the SCFs. The authors concluded that the sample size limited
A Phase III studied by Skarlos et al. (2001) described their results, but cautioned that omission of ENI on
treatment to gross tumor, hilum, mediastinum, and the basis of CT scans in patients with LS-SCLC
involved SCF, as did the Japan Clinical Oncology resulted in a higher than expected rate of isolated
Group Phase III study published by Takada et al. nodal failures in the ipsilateral SCF. In the 2006
(2002) with the initial field in the sequential arm Phase II study by Baas et al. (2006) the TRT target
based on the pre-CHT tumor volume. In 2006, the volume for irradiation was planned to start within one
most recent Phase III study in LS-SCLC was pub- week after the start of the second cycle of CHT and
lished and was a replication of the early versus late included the primary tumor and all clinical and
initiation of RT with CHT by Murray et al. (1993). radiologically involved lymph nodes with a short axis
The field size described by Spiro et al. (2006) was diameter of [1 cm; this was termed ‘‘involved field
based on the pre-CHT tumor and used the target irradiation’’ (Baas et al. 2006). A PET scan was not
definitions of the Murray trial. performed. The authors reported an in-feld recurrence
Relatively recent trials have remained conservative rate of 24%. Out-of-field failures were only seen in 2
in their approach to MLN inclusion in the target. In patients. Belderbos et al. (2007) compared the failure
Bogart et al. (2004) phase II trial of dose escalation in patterns in this work by Baas et al. (2006) with those
SCLC planning was based on volumes from the of de Ruysscher study (2006) and noted that isolated
restaging chest CT obtained after induction CHT, with SCF failures were seen in both studies. Since it had
the GTV including residual lung tumor after induction been reported that routine ultrasound of the supra-
CHT, but with involved lymph node regions (both pre- clavicular area has improved the clinical (CT) staging
CHT and post-CHT) (Bogart et al. 2004). A 2007 report of SCLC patients (van Overhagen et al. 2004),
by Schild et al. on the results of a Phase II study of high- Belderbos et al. (2007) suggested that this test be
dose radiotherapy with concurrent chemotherapy incorporated in the assessment of LS-SCLC in whom
(Schild et al. 2007) had the target initially treated with non-ENI RT fields are being contemplated. van Loon
anteroposterior-posteroanterior fields that included et al. (vl e 42) provided results from a planning study
primary tumor with ipsilateral hilar, mediastinal, and incorporating FDG-PET in the TRT planning for
SCF nodes. Oblique fields were then tailored to include target definition and reported 24% of treatment plans
primary tumor, ipsilateral hilum, and SCF nodes if were changed compared to CT-based planning, with
initially involved, and a ‘‘reduced’’ volume of the both increases and decreases observed in GTV. More
mediastinum, although the parameters for reduction recently, van Loon and her colleagues published
were unspecified. Achieving small treatment portals results from a series of 60 patients treated with con-
was a study goal and so fields were designed after the current CHT and hyperfractionated TRT [45 Gy],
fourth and fifth cycles of CHT. with PET scan based selective nodal irradiation
Studies specifically addressing ENI issues have (van Loon et al. 2010). They observed a low rate of
been relatively recent and have come from extensive isolated nodal failures (3%). This was in contrast
work in the Netherlands. The first clinical study to with the findings from their aforementioned study of
208 G. M. M. Videtic

CT-based selective nodal irradiation, which resulted not identified by the conventional staging methods.
in a higher percentage of isolated nodal failures (11%) In 5 patients with limited disease, PET detected
(de Ruysscher et al. 2006). A recent (2010) retro- additional metastatic foci: ipsilateral pulmonary
spective study from the US by Watkins et al. (2010) metastasis (n = 1) and contralateral mediastinal
of 52 patients treated with hyperfractionated TRT (n = 1), contralateral supraclavicular (n = 2), and
starting at CHT cycle 1 or 2 and with involved MLNs contralateral cervical (n = 1) lymph node metastases.
defined by CT and/or PET criteria, showed that Several prospective and retrospective studies have
involved-field TRT did not appear to have an adverse noted the improved sensitivity and specificity of PET
impact on anticipated patterns of failure or survival over CT with respect to identifying mediastinal nodal
(Watkins et al. 2010). abnormalities, but most of these reports did not vali-
date clinical results with pathologic sampling, nor did
they discuss implications for therapy.
5 SCLC Target Definition In summary, PET imaging contributes substan-
and FDG-PET tially to better identification of tumor burden, and can
definitely influence RT treatment plans. A major
The previously noted Phase II studies of ENI in weakness of all these studies is the absence of histo-
LS-SCLC distinguished the use of CT imaging for logic verification of PET and CT findings and none of
defining abnormal MLNs from that obtained from the cited studies were correlated with failure patterns.
PET imaging. In NSCLC, it has been shown that PET However, it remains a powerful tool for more accu-
has a higher sensitivity, specificity, and accuracy for rately staging and defining gross tumor.
detecting tumor involvement in MLNs than CT
imaging (van Baardwijk et al. 2006). However, the
gold standard for defining the presence and extent of 6 SCLC Target Definition
MLN metastases in NSCLC has long been, and in Extensive Disease
remains, mediastinoscopy (Rusch 2005). Validation
of PET-defined nodal targets in RT planning for Most clinicians conventionally consider the role of
NSCLC against the pathologic standard, however, is a RT in ES-SCLC as essentially palliative. However,
little-investigated area. The question of PET based the role of RT in ES-SCLC has regained interest
imaging and pathologic validation in SCLC as it because of a recent randomized study of PCI by
applies to RT planning has not been studied. Slotman et al. (2007) in ES-SCLC patients with a
There are a number of recent publications report- response after CHT that showed a clear benefit in
ing on the utility of PET in the staging of SCLC. Two terms of lower brain relapses and higher overall sur-
studies are of interest with respect to RT planning. vival. These data recall a not-yet-replicated phase III
Bradley et al. (2004) prospectively performed PET study on the role of thoracic RT in favorable ES-
scanning on 24 patients determined by conventional SCLC patients by Jeremic et al. (1999). In that study,
staging to have LS-SCLC. PET identified unsuspected accelerated hyperfractionated RT and concurrent low-
regional nodal metastases in 6 (25%) of 24 patients dose daily CHT were instituted after three induction
compared with CT: 1 patient with N1 disease on CT cycles of cisplatin-etoposide (PE) and compared with
was found to have N2 disease by PET; 5 patients with CHT alone. PCI was offered to responders in both
clinical N2 disease on CT were found to have N3 groups. The target volume in the TRT group included
disease. As in other studies, none of the PET findings all gross disease and the ipsilateral hilum with a 2 cm
was confirmed histologically. The RT plan, however, margin and the entire mediastinum with a 1 cm
was significantly altered to include the PET-positive/ margin. Both SCF were routinely irradiated. Although
CT-negative nodes within the high-dose region in TRT improved local control and survival, there was
each of these patients. In a retrospective study by no difference between combined RT-CHT and CHT
Kamel et al. (2003), PET scans had an impact on RT alone group in terms of bronchopulmonary toxicity.
in 8 patients (19%). In 5 patients (12%), PET scans This important study merits replication, employing
resulted in a change of RT field and volume after contemporary RT technologies and concepts for tar-
identifying additional active tumor foci, which were get delineation.
The Radiation Target in Small-Cell Lung Cancer 209

different approaches to target delineation, with the


7 SCLC Target Definition in Current former having a more rigorous non-ENI approach and
Trials each having an impact factor from the timing of CHT
administration.
The European Organization for Research and Treat-
ment of Cancer (EORTC) is currently running a
Phase III trial in LS-SCLC comparing two RT doses 8 Conclusions
(European Organisation for Research and Treatment
of Cancer (EORTC) trial 2011). CONVERT (Con- The criteria for defining the appropriate target when
current once-daily VErsus twice-daily RadioTherapy) treating a SCLC patient remains an active area of
is a Phase III trial in which patients with LS-SCLC investigation. Two ongoing large randomized clinical
are randomized to either twice-daily thoracic radio- trials should provide the most up-to-date evidence to
therapy (45 Gy in 30 fractions starting with the sec- guide radiation oncologists in optimal planning of RT
ond cycle of PE) or to a higher dose of conventional delivery to the tumor. It is likely that technological
radiation (66 Gy in 33 daily fractions over 6.5 weeks advances in RT delivery may have their greatest
starting with the second cycle of PE). The primary impact with respect to normal tissue toxicities. In the
end point is overall survival. ENI is explicitly not absence of strong evidence supporting omission of
employed. TRT is to be started within 3 weeks of ENI, clinicians must use thoughtful clinical judgment,
planning. The GTV is defined as all identifiable tumor integrating a number of tools such as PET imaging
and involved LNs (defined on CT as nodes [1 cm in along with appropriate interpretation of the evidence
short axis). The CTV comprises the GTV with a to guide their treatment planning, with an emphasis
0.5 cm margin. The PTV includes the CTV with a on a balance between increase of failure risk and
1.5 cm margin superiorly and inferiorly and 1 cm maximal reduction of treatment-related toxicities to
margin laterally. If PET scans are available for stag- support improvements in outcomes.
ing, the GTV is required to include PET-positive LNs.
Also currently ongoing is a randomized three-arm
dose comparison trial in LS-SCLC initiated by the
major North American collaborative groups (CALGB References
30610/RTOG 0538) (Phase III randomized study of
three different thoracic radiotherapy regimens in Aristei C, Falcinelli L, Palumbo B, Tarducci R (2010) PET and
PET-CT in radiation treatment planning for lung cancer.
patients with limited-stage small-cell lung cancer Expert Rev Antic Ther 10:571–584
receiving cisplatin and etoposide. 2011). The primary Arriagada R, Pellae-Cosset B, Ladron de Guevara JC, el Bakry H,
objective of this study is to determine whether Benna F, Martin M, de Cremoux H, Baldeyrou P, Cerrina ML,
administering concurrent PE starting at cycle 1 or 2 Le Chevalier T (1991) Alternating radiotherapy and chemo-
therapy schedules in limited small cell lung cancer: analysis
with high-dose TRT as 70 Gy (2 Gy once-daily over of local chest recurrences. Radiother Oncol 20:91–98
7 weeks) or 61.2 Gy (1.8 Gy once-daily for 16 days Auperin A, Arriagada R, Pignon JP, Le Pechoux C, Gregor A,
followed by 1.8 Gy twice-daily for 9 days), will Stephens RJ et al (1999) Prophylactic cranial irradiation for
improve median and 2-year survival compared with patients with small-cell lung cancer in complete remission.
Prophylactic cranial irradiation overview collaborative
45 Gy (1.5 Gy twice-daily over 3 weeks) in patients group. N Engl J Med 341:476–484
with LS-SCLC. In this study, the GTV is defined as Baas P, Belderbos JS, Senan S, Kwa HB, van Bochove A, van
the primary tumor and clinically positive LNs seen Tinteren H, Burgers JA, van Meerbeeck JP (2006) Concur-
either on the pretreatment CT ([1 cm short axis rent chemotherapy (carboplatin, paclitaxel, etoposide) and
involved-field radiotherapy in limited stage small cell lung
diameter) or on pretreatment PET scan (SUV [ 3). cancer: a Dutch multicenter phase II study. Br J Cancer
There will be two CTVs, 1 or 2, which will be used as 94:625–630
a function of the arm to which the patient is ran- Belderbos J, Baas P, Senan S (2007) Reply: patterns of nodal
domized. CTV-1 will include the GTV and the ipsi- recurrence after omission of elective nodal irradiation for
limited-stage small-cell lung cancer. British J Cancer
lateral hilum and CTV-2 will consist of a revised 97:276
GTV-only based on CHT response. Thus CONVERT Bleehen NM, Bunn PA, Cox JD, Dombernowsky P, Fox RM,
and the US Intergroup study are employing slightly Høst H, Joss R, White JE, Wittes RE (1983) Role of
210 G. M. M. Videtic

radiation therapy in small cell anaplastic carcinoma of the International Commission on Radiation Units and Measure-
lung. Cancer Treat Rep 67:11–19 ments (1993) Prescribing, recording, and reporting photon
Bogart JA, Herndon JE 2nd, Lyss AP, Watson D, Miller AA, beam therapy, ICRU Report 50, MD, USA
Lee ME, Turrisi AT, Green MR, Cancer and Leukemia International Commission on Radiation Units and Measure-
Group B study 39808 (2004) 70 Gy thoracic radiotherapy is ments (1999) Prescribing, recording, and reporting photon
feasible concurrent with chemotherapy for limited-stage beam therapy (Supplement to ICRU Report 50). ICRU
small-cell lung cancer: analysis of cancer and leukemia Report 62, MD, USA
group B study 39808. Int J Radiat Oncol Biol Phys 59:460– Jeremic B (2006) Timing of concurrent radiotherapy and
468 chemotherapy in limited-disease small-cell lung cancer:
Bradley JD, Dehdashti F, Mintun MA, Govindan R, Trinkaus ‘‘Meta-analysis of meta-analyses’’. Int J Radiat Oncol Biol
K, Siegel BA (2004) Positron emission tomography in Phys 64:981–982
limited-stage small-cell lung cancer: a prospective study. Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1997)
J Clin Oncol 22:3248–3254 Initial versus delayed accelerated hyperfractionated radia-
Brodin O, Rikner G, Steinholtz L, Nou E (1990) Local failure tion therapy and concurrent chemotherapy in limited small-
in patients treated with radiotherapy and multidrug chemo- cell lung cancer: a randomized study. J Clin Oncol 15:893–
therapy for small cell lung cancer. Acta Oncol 29:739–746 900
Bunn PA Jr, Lichter AS, Makuch RW, Cohen MH, Veach SR, Jeremic B, Shibamoto Y, Nikolic N, Milicic B, Milisavljevic S,
Matthews MJ, Anderson AJ, Edison M, Glatstein E, Minna Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999)
JD et al (1987) Chemotherapy alone or chemotherapy with Role of radiation therapy in the combined-modality treat-
chest radiation therapy in limited stage small cell lung ment of patients with extensive disease small-cell lung
cancer. A prospective, randomized trial. Ann Intern Med cancer: a randomized study. J Clin Oncol 17:2092–2099
106:655–662 Kamel EM, Zwahlen D, Wyss MT, Stumpe KD, von Schulthess
Cooper S, Spiro SG (2006) Small cell lung cancer: treatment GK, Steinert HC (2003) Whole-body (18) F-FDG PET
review. Respirology 11:241–248 improves the management of patients with small cell lung
Curran WJ Jr (2001) Therapy of limited stage small cell lung cancer. J Nucl Med 44:1911–1917
cancer. Cancer Treat Res 105:229–252 Kies MS, Mira JG, Crowley JJ, Chen TT, Pazdur R, Grozea PN,
de Ruysscher D, Vansteenkiste J (2000) Chest radiotherapy in Rivkin SE, Coltman CA Jr, Ward JH, Livingston RB (1987)
limited-stage small cell lung cancer: facts, questions, Multimodal therapy for limited small-cell lung cancer: a
prospects. Radiother Oncol 55:1–9 randomized study of induction combination chemotherapy
de Ruysscher D, Bremer RH, Koppe F, Wanders S, van Haren with or without thoracic radiation in complete responders;
E, Hochstenbag M, Geeraedts W, Pitz C, Simons J, ten and with wide-field versus reduced-field radiation in partial
Velde G, Dohmen J, Snoep G, Boersma L, Verschueren T, responders: a southwest oncology group study. J Clin Oncol
van Baardwijk A, Dehing C, Pijls M, Minken A, Lambin P 5:592–600
(2006) Omission of elective node irradiation on basis of CT- Kong FM, Gaspar LE, Komaki R, Sun A, Bonner JA, Choy H,
scans in patients with limited disease small cell lung cancer: Wang L, Morris D, Sandler HM, Movsas B (2007) Patterns
a phase II trial. Radiother Oncol 80:307–312 of practice in radiation dose prescription and treatment
Emami B (1996) Three-dimensional conformal radiation therapy planning for patients with lung cancer among members of
in bronchogenic carcinoma. Semin Radiat Oncol 6:92–97 American society of therapeutic radiology and oncology. Int
European Organisation for Research and Treatment of Cancer J Radiat Oncol Biol Phys 69(Suppl 1):S483
(EORTC) trial (2007) A randomized controlled trial of Lee CB, Morris DE, Fried DB et al (2006) Current and
concurrent chemo-radiotherapy comparing twice-daily and evolving treatment options for limited stage small cell lung
once-daily radiotherapy schedules in patients with limited cancer. Curr Opin Oncol 18:162–172
stage small celllung cancer (SCLC) and good performance Liengswangwong V, Bonner JA, Shaw EG, Foote RL, Frytak S,
status. http://www.christie.nhs.uk/research/themes/convert/ Eagan RT, Jett JR, Richardson RL, Creagan ET, Su JQ
default.aspx. Accessed 13 Mar 2011 (1994) Limited-stage small-cell lung cancer: patterns of
Faivre-Finn C, Lee LW, Lorigan P, West C, Thatcher N intrathoracic recurrence and the implications for thoracic
(2005a) Thoracic radiotherapy forlimited-stage small-cell radiotherapy. J Clin Oncol 12:496–502
lung cancer: controversies and future developments. Clin Mira JG, Livingston RB (1980) Evaluation and radiotherapy
Oncol (R Coll Radiol) 17:591–598 implications of chest relapse patterns in small cell lung
Faivre-Finn C, Lorigan P, West C, Thatcher N (2005b) carcinoma treated with radiotherapy-chemotherapy: study
Thoracic radiation therapy for limited-stage small-cell lung of 34 cases and review of the literature. Cancer 46:2557–
cancer: unanswered questions. Clin Lung Cancer 7:23–29 2565
Fried DB, Morris DE, Poole C, Rosenman JG, Halle JS, Mira JG, Livingston RB, Moore TN, Chen T, Batley F,
Detterbeck FC, Hensing TA, Socinski MA (2004) System- Bogardus CR Jr, Considine B Jr, Mansfield CM, Schlosser J,
atic review evaluating the timing of thoracic radiation Seydel HG (1982) Influence of chest radiotherapy in
therapy in combined modality therapy for limited-stage frequency and patterns of chest relapse in disseminated
small-cell lung cancer. J Clin Oncol 22:4785–4793 small cell lung carcinoma. A southwest oncology group
Huncharek M, McGarry R (2004) A meta-analysis of the timing study. Cancer 50:1266–1272
of chest irradiation in the combined modality treatment of Murray N, Coy P, Pater JL, Hodson I, Arnold A, Zee BC, Payne
limited-stage small cell lung cancer. Oncologist 9:6665– D, Kostashuk EC, Evans WK, Dixon P et al (1993)
6672 Importance of timing for thoracic irradiation in the
The Radiation Target in Small-Cell Lung Cancer 211

combined modality treatment of limited-stage small-cell EORTC radiation oncology group and lung cancer group.
lung cancer. The national cancer institute of Canada clinical Prophylactic cranial irradiation in extensive small-cell lung
trials group. J Clin Oncol 11:336–344 cancer. N Engl J Med 357:664–672
Perez CA, Krauss S, Bartolucci AA, Durant JR, Lowenbraun S, Socinski MA, Bogart JA (2007) Limited-stage small-cell lung
Salter MM, Storaalsi J, Kellermeyer R, Comas F (1981) cancer: the current status of combined-modality therapy.
Thoracic and elective brain irradiation with concomitant or J Clin Oncol 25:4137–4145
delayed multiagent chemotherapy in the treatment of Spiro SG, James LE, Rudd RM, Trask CW, Tobias JS, Snee M,
localized small cell carcinoma of the lung: a randomized Gilligan D, Murray PA, Ruiz de Elvira MC, O’Donnell KM,
prospective study by the southeastern cancer study group. Gower NH, Harper PG, Hackshaw AK, London Lung
Cancer 47:2407–2413 Cancer Group (2006) Early compared with late radiotherapy
Perez CA, Einhorn L, Oldham RK, Greco FA, Cohen HJ, in combined modality treatment for limited disease small-
Silberman H, Krauss S, Hornback N, Comas F, Omura G, cell lung cancer: a London lung cancer group multicenter
et al (1984) Randomized trial of radiotherapy to the thorax randomized clinical trial and meta-analysis. J Clin Oncol
in limited small-cell carcinoma of the lung treated with 24:3823–3830
multiagent chemotherapy and elective brain irradiation: a Tada T, Minakuchi K, Koda M, Masuda N, Matsui K, Kawase
preliminary report. J Clin Oncol 2:1200–1208 I, Nakajima T, Nishioka M, Fukuoka M, Kozuka T (1998)
Perry MC, Eaton WL, Propert KJ, Ware JH, Zimmer B, Limited-stage small cell lung cancer: local failure after
Chahinian AP, Skarin A, Carey RW, Kreisman H, chemotherapy and radiation therapy. Radiology 208:511–
Faulkner C et al (1987) Chemotherapy with or without 515
radiation therapy in limited small-cell carcinoma of the Takada M, Fukuoka M, Kawahara M, Sugiura T, Yokoyama A,
lung. N Engl J Med 316:912–918 Yokota S, Nishiwaki Y, Watanabe K, Noda K, Tamura T,
Phase III randomized study of three different thoracic radio- Fukuda H, Saijo N (2002) Phase III study of concurrent
therapy regimens in patients with limited-stage small cell versus sequential thoracic radiotherapy in combination with
lung cancer receiving cisplatin and etoposide. cisplatin and etoposide for limited-stage small-cell lung
https://www.ctsu.org/public/data/protocols/CALGB/ cancer: results of the Japan clinical oncology group study
CALGB-30610/pfs.pdf. Accessed 13 Mar 2011 9104. J Clin Oncol 20:3054–3060
Pignon JP, Arriagada R, Ihde DC, Johnson DH, Perry MC, Turrisi AT 3rd, Kim K, Blum R, Sause WT, Livingston RB,
Souhami RL, Brodin O, Joss RA, Kies MS, Lebeau B et al Komaki R, Wagner H, Aisner S, Johnson DH (1999) Twice-
(1992) A meta-analysis of thoracic radiotherapy for small- daily compared with once-daily thoracic radiotherapy in
cell lung cancer. N Engl J Med 327:1618–1624 limited small-cell lung cancer treated concurrently with
Pijls-Johannesma M, de Ruysscher D, Vansteenkiste J, Kester cisplatin and etoposide. N Engl J Med 340:265–2671
A, Rutten I, Lambin P (2007) Timing of chest radiotherapy van Baardwijk A, Baumert BG, Bosmans G, van Kroonenburgh
in patients with limited stage small cell lung cancer: a M, Stroobants S, Gregoire V, Lambin P, de Ruysscher D
systematic review and meta-analysis of randomised con- (2006) The current status of FDG-PET in tumour volume
trolled trials. Cancer Treat Rev 33:461–473 definition in radiotherapy treatment planning. Cancer Treat
Rusch VW (2005) Mediastinoscopy: an endangered species? Rev 32:245–260
J Clin Oncol 23:8283–8285 van Loon J, de Ruysscher D, Wanders R, Boersma L, Simons J,
Samson DJ, Seidenfeld J, Simon GR, Turrisi AT 3rd, Bonnell Oellers M, Dingemans AM, Hochstenbag M, Bootsma G,
C, Ziegler KM, Aronson N American College of Chest Geraedts W, Pitz C, Teule J, Rhami A, Thimister W, Snoep
Physicians (2007) Evidence for management of small cell G, Dehing-Oberije C, Lambin P (2010) Selective nodal
lung cancer: ACCP evidence-based clinical practice guide- irradiation on basis of (18)FDG-PET scans in limited-
lines, 2nd edn. Chest 132(3 Suppl):314S–323S disease small-cell lung cancer: a prospective study. Int J
Schild SE, Bonner JA, Hillman S et al (2007) Results of a phase Radiat Oncol Biol Phys 77:329–336
II study of high-dose thoracic radiation therapy with van Overhagen H, Brakel K, Heijenbrok MW, van Kasteren JH,
concurrent cisplatin and etoposide in limited-stage small- van de Moosdijk CN, Roldaan AC, van Gils AP, Hansen BE
cell lung cancer (NCCTG 95–20-53). J Clin Oncol (2004) Metastases in supraclavicular lymph nodes in lung
25:3124–3129 cancer: assessment with palpation, US, and CT. Radiology
Senan S, de Ruysscher D (2005) Critical review of PET-CT for 232:75–80
radiotherapy planning in lung cancer. Crit Rev Oncol Videtic GM, Belderbos JS, Spring Kong FM, Kepka L,
Hematol 56:345–351 Martel MK, Jeremic B (2008) Report from the interna-
Skarlos DV, Samantas E, Briassoulis E, Panoussaki E, Pavlidis tional atomic energy agency (IAEA) consultants’ meeting
N, Kalofonos HP, Kardamakis D, Tsiakopoulos E, Kosmidis on elective nodal irradiation in lung cancer: small-cell
P, Tsavdaridis D, Tzitzikas J, Tsekeris P, Kouvatseas G, lung cancer (SCLC). Int J Radiat Oncol Biol Phys
Zamboglou N, Fountzilas G (2001) Randomized compari- 72:327–334
son of early versus late hyperfractionated thoracic irradia- Warde P, Payne D (1992) Does thoracic irradiation improve
tion concurrently with chemotherapy in limited disease survival and local control in limited-stage small-cell
small-cell lung cancer: a randomized phase II study of the carcinoma of the lung? A meta-analysis. J Clin Oncol
Hellenic cooperative oncology group (HeCOG). Ann Oncol 10:890–895
12:1231–1238 Watkins JM, Wahlquist AE, Zauls AJ, Shirai K, Garrett-Mayer
Slotman B, Faivre-Finn C, Kramer G, Rankin E, Snee M, E, Aguero EG, Silvestri GA, Sherman CA, Sharma AK
Hatton M, Postmus P, Collette L, Musat E, Senan S (2007) (2010) Involved-field radiotherapy with concurrent
212 G. M. M. Videtic

chemotherapy for limited-stage small-cell lung cancer: of the lung: preliminary report of a southwest oncology
disease control, patterns of failure and survival. J Med group study. Cancer 50:1084–1090
Imaging Radiat Oncol 54:483–489 Work E, Nielsen OS, Bentzen SM, Fode K, Palshof T (1997)
White JE, Chen T, McCracken J, Kennedy P, Seydel HG, Randomized study of initial versus late chest irradiation
Hartman G, Mira J, Khan M, Durrance FY, Skinner O combined with chemotherapy in limited-stage small-cell
(1982) The influence of radiation therapy quality control on lung cancer. Aarhus lung cancer group. J Clin Oncol
survival, response and sites of relapse in oat cell carcinoma 15:3030–3037
Radiation Sensitizers
Anthony M. Brade and Zishan Allibhai

Contents Abstract
Discovery of effective radiosensitization strategies
1 Radiation Sensitizers ............................................... 213 that improve the therapeutic ratio for patients with
2 Oxygen ...................................................................... 214 lung cancer has been a goal of researchers and an
2.1 Hyperbaric Oxygen.................................................... 214 area of vigorous investigation for the past several
2.2 Carbogen .................................................................... 215 decades. A pure radiosensitizer is a drug, a
2.3 Efaproxiral ................................................................. 215
modality of therapy or an intervention that, on its
2.4 Red Blood Cell Transfusion ..................................... 215
2.5 Erythropoietin ............................................................ 216 own, lacks direct anti-tumor activity but enhances
the cytotoxicity of radiotherapy when employed in
3 Targeting Hypoxic Cells ......................................... 216
3.1 Hypoxic Cell Radiosensitizers .................................. 216 combination. In this chapter we outline the previ-
3.2 Bioreductive Drugs (Hypoxic Cell Cytotoxins) ....... 217 ous and ongoing attempts to radiosensitize lung
cancers through improved tumor oxygenation,
4 Boron Neutron Capture Theory ............................ 217
augmentation of the effectiveness of radiotherapy
5 DNA Repair Inhibitors ........................................... 217 in hypoxic tumor cells, and use of drugs that
5.1 AKT Pathway Inhibition ........................................... 217
5.2 Mitogen-Activated Protein Kinase Inhibition .......... 218 modulate with DNA repair and apoptosis. To date,
5.3 HDAC-Inhibitors ....................................................... 218 no pure radiosensitization strategy has established
6 Apoptosis Modulating Agents ................................ 219
itself in standard practice but the current research
6.1 Bcl-2 Inhibitors.......................................................... 219 suggests that this field continues to hold great
6.2 Cyclooxygenase COX-2-Inhibitors ........................... 219 promise for the improvement of outcome in
References.......................................................................... 219 patients with lung cancer.

1 Radiation Sensitizers

Although radiotherapy has traditionally played a


major role in the treatment of non-small cell lung
cancers (NSCLC), its effectiveness had been limited
A. M. Brade (&)  Z. Allibhai by an unfavourable therapeutic ratio, whereby it is
Department of Radiation Oncology, challenging to deliver optimal doses of radiation to
Princess Margaret Hospital,
tumors without excessive normal tissue toxicity.
5-912 610 University Avenue, Toronto,
ON M5G 2M9, Canada Recently, a number of technological advances in
e-mail: Anthony.brade@rmp.uhn.on.ca both imaging and radiation delivery have further
A. M. Brade improved our ability to spare normal tissue. A further
University of Toronto, Toronto, Canada approach to enhancing the therapeutic ratio is to

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 213
DOI: 10.1007/174_2011_278, Ó Springer-Verlag Berlin Heidelberg 2011
214 A. M. Brade and Z. Allibhai

selectively increase the radiosensitivity of the target described by the oxygen-enhancement ratio (OER)
tumor. The advantages of this approach are poten- which quantifies the relative dose required to produce
tially significant given that the presumed radioresis- a certain biologic effect under hypoxic conditions
tance of NSCLC has long been a major obstacle to the versus that required to achieve the same effect under
efficacy of RT. aerobic conditions.
A pure radiosensitizer is a drug, a modality of The presence of a hypoxic cell subpopulation is
therapy or an intervention that on its own lacks direct a common feature of solid tumors, and although the
anti-tumor activity but enhances the cytotoxicity of extent of hypoxia varies widely from tumor to
radiotherapy when employed in combination. Many tumor and within tumors, it contributes significantly
chemotherapeutic agents (e.g. platins, taxanes, and to radioresistance. While the clinical effect of
topoisomerase modulators) enhance the cytotoxicity of hypoxia on cancer outcomes has been best dem-
radiotherapy when the two modalities are combined but onstrated in the setting of head and neck cancers
as they generally effect some degree of anti-tumor (Becker et al. 1998), cervical cancer (Hockel et al.
activity when administered as single agents, they 1993), and soft tissue sarcomas (Brizel et al. 1995),
are therefore not considered true radiosensitizers. it has also been shown in NSCLC. For example,
Similarly, newer classes of drugs have been developed mean hemoglobin levels were associated with the
to exploit tumor-specific mutations or target-specific degree of pathologic response seen following neo-
molecular-based alterations within the tumor or its adjuvant chemoradiation (Robnett et al. 2002)
microenvironment. Some of these also augment the while low (Laurie et al. 2006) or declining
efficacy of radiotherapy and have single agent efficacy (MacRae et al. 2002) hemoglobin levels during the
against lung cancer and are thus not pure radiosensi- course of chemoradiation have been correlated with
tizers. For example, tyrosine kinase inhibitors of the worse overall survival. Tumor hypoxia in NSCLC
epidermal growth factor receptor such as erlotinib and has also been correlated with an increased inci-
gefitinib have shown impressive enhancement in tumor dence of distant metastasis (Le et al. 2007) and
models when combined with radiation treatment poorer overall prognosis (Eschmann et al. 2005).
(Chinnaiyan et al. 2005; Tanaka et al. 2008; Park et al. Hypoxia has been demonstrated to trigger the
2010) in addition to improving outcome as single downregulation of key DNA repair pathways,
agents in patients with NSCLC (Tsao et al. 2005), leading to genetic instability. Thus, tumor hypoxia
particularly in patients bearing specific mutations in the is a key driver of metastatic spread and treatment
EGFR gene (Mok et al. 2009). Other examples include resistance (Brizel et al. 1996; Brown and Gaccia
the use of agents that inhibit vascular endothelial 1998).
growth factor (VEGF) (Manegold et al. 2008), inhibi- Considerable effort has been focused on discov-
tors of anaplastic lymphoma kinase (ALK), and ering interventions by which tumor hypoxia can be
inhibitors of mammalian target of rapamycin (mTOR) reduced. These have included artificial means of
(Nagata et al. 2010). Combinations of radiotherapy increasing hemoglobin oxygenation levels as well
with chemotherapy or molecularly-targeted drugs in as measures to optimize the hemoglobin levels
lung cancer are detailed elsewhere in this volume. This themselves.
chapter will focus on pure-radiosensitizing strategies
and their potential relevance to the treatment of lung 2.1 Hyperbaric Oxygen
cancer.
A hyperbaric environment allows greater solubility of
oxygen within plasma and hemoglobin saturation,
2 Oxygen thus maximizing oxygen delivery to hypoxic tumor
cells. While randomized studies conducted in patients
One of the earliest radiosensitizers to be identified and with head and neck cancers (Henk et al. 1977) as well
studied was oxygen. Its presence increases the yield, as cervical cancer (Watson et al. 1978) had shown
variety, and lifetime of the radical species formed improvements in local control and/or survival, a
secondary to tumor/tissue irradiation. The impact of number of trials studying HBO in various other
oxygen on radiation effect is mathematically cancer sites failed to show a benefit. Furthermore,
Radiation Sensitizers 215

there exist a number of logistical issues in both the 2.3 Efaproxiral


availability and administration of HBO during a
standard course of fractionated radiotherapy. As a Efaproxiral (RSR-13) is an allosteric effector of
result, the role of HBO in clinical practice has been hemoglobin which enhances its oxygen-carrying
limited to the repair of damaged tissues following RT capacity, effectively increasing the delivery of oxygen
where it has been shown to limit and in some cases to otherwise hypoxic regions. Phases I and II studies
reverse the process of radiation damage (Bennett et al. demonstrated that this compound is safe and effective
2005; Bui et al. 2004). in improving tumor oxygenation and potentiating
radiation effect. The radiation enhancing allosteric
compounds of hypoxic brain metastases (REACH)
2.2 Carbogen trial was a phase III study designed to evaluate the
addition of efaproxiral to whole brain radiotherapy in
Carbogen is a mixture of oxygen and carbon dioxide patients with brain metastasis (Suh et al. 2006). No
(typically 95% O2 to 5% CO2) and it has a theoretical difference in overall survival was found for the entire
advantage over pure oxygen since the increased level group, over half of whom had metastatic NSCLC. An
of carbon dioxide triggers a rightward shift of the unplanned subset analysis did however show that in
oxyhemoglobin curve, thus facilitating the release of the subset of patients with metastatic breast cancer
O2 into areas of hypoxia (Rubin et al. 1979). (20% of patients), the addition of efaproxiral
Carbogen has also been used in combination improved survival as well as quality of life (Scott
with agents that enhance tumor blood flow, for e.g. et al. 2007).
nicotinamide, thus providing a synergistic effect. A phase II trial was designed to test the efficacy
This combination of agents was studied in a phase and safety of efaproxiral given concurrently with
II trial from the Netherlands which found that the radiation following induction chemotherapy for
use of ARCON (accelerated radiotherapy with locally advanced NSCLC. This combination yielded
carbogen and nicotinamide) yielded high control an overall response rate of 75% and a very encour-
rates in laryngeal cancer (Kaanders et al. 1998). aging median survival of 20.6 months, while the rates
While data regarding late toxicity and outcome data of severe toxicity were relatively low (Choy et al.
are not yet mature, the subsequent phase III study 2005). These encouraging results prompted the pro-
comparing ARCON versus accelerated radiation posal of a phase III study, however, this trial was
only demonstrated a mild increase in confluent never launched (Choy, personal correspondence).
mucositis (Janssens et al. 2011). This is consistent
with the results from a phase III study in locally
advanced bladder cancer where the addition of 2.4 Red Blood Cell Transfusion
ARCON to RT improved both local control and
overall survival without significantly increasing late Transfusion represents a potentially direct means of
morbidity (Hoskin et al. 2010). improving oxygen delivery to tumors in anemic
In regard to lung cancer, a similar phase I/II study patients. An important study evaluating transfusion to
was launched in Switzerland to investigate the effects counter anemia was undertaken in cervical cancer
of adding carbogen and/or nicotinamide to acceler- patients at Princess Margaret Hospital. This trial
ated radiation for locally advanced NSCLC staged suggested that transfusions might improve outcomes
IIIA or IIIB. There was no difference in radiologic in patients who were anemic while undergoing
response or time to progression between the three definitive radiotherapy (Fyles et al. 1998).
groups, and no increase in radiotherapy related The systemic administration of hemoglobin vesicle
adverse events. However, a significant proportion of (HbV), which serves as an artificial oxygen carrier,
patients in the two groups that received nicotinamide represents a recently developed alternative to red cell
developed significant (CG2) emesis which was shown transfusion. Animal studies of HbV have demon-
to adversely affect their quality of life (Bernier et al. strated that it increases oxygen tension within tumor
1999), thus this strategy is unlikely to be pursued tissue and that this positively affects the tumor’s
further in this disease. response to radiation (Yamamoto et al. 2009).
216 A. M. Brade and Z. Allibhai

This was the first study to test tumor oxygenation their SERs were also reduced and the two large phase
using a liposome-type artificial oxygen carrier, and III trials studying etanidazole in locally advanced
further studies are anticipated. head and neck carcinomas did not yield any
improvements in outcome over conventionally frac-
tionated radiation alone (Lee et al. 1995; Eschwège
2.5 Erythropoietin et al. 1997).
Nimorazole was also found to have a lower SER
Erythropoietin (EPO) is a glycoprotein hormone that than misonidazole, however, significantly greater
controls erythropoiesis through its interactions with doses could be tolerated since it was far less toxic
the erythropoietin receptor (EpoR) found on bone than any of its predecessors (Overgaard 1994). This
marrow stem cells. Human recombinant erythropoie- agent was subsequently evaluated in the Danish Head
tin (and similar exogenous agents) represent another and Neck Cancer (DAHANCA) 5 study. This phase
method by which hemoglobin levels may be increased, III trial included 422 patients with supraglottic and
and a number of trials have studied the effect of EPO pharyngeal carcinomas. The addition of nimorazole
in the management of anemic cancer patients. improved 5 year locoregional control from 33 to 49%
Although encouraging results were found in anemic (P \ 0.002) and cancer-specific survival improved
lung cancer patients undergoing chemotherapy from 41 to 52% (P = 0.01). Importantly, there was no
(Vansteenkiste et al. 2002), subsequent trials in breast increase in severe late radiation morbidity (Overgaard
and head and neck cancer patients actually suggested et al. 1998). This study has resulted in nimorazole
that these agents worsened outcomes (Henke et al. being incorporated into standard practice in Denmark.
2003; Leyland-Jones 2003). A subsequent meta- However, the routine use of radiosensitizers for head
analysis of 9,353 patients from 53 studies studying the and neck cancer has not generally been adopted out-
effects of epoetin or darbopoetin demonstrated an side Denmark, likely due to the fact that other phase
increased risk of thrombo-embolic events as well as III trials yielded negative or inconclusive results.
increased mortality from the use of these agents It should be noted that many of these other studies
(Bohlius et al. 2009). were underpowered and used earlier generations of
radiosensitizing agents that were less effective and
more toxic (Ang 2010).
3 Targeting Hypoxic Cells A meta-analysis of 82 different trials involving
HBO and hypoxic-cell radiosensitizers showed a
3.1 Hypoxic Cell Radiosensitizers 4.6% improvement in local tumor control (Kaanders
et al. 2004). Although few of these trials focused on
Hypoxic cell radiosensitizers are compounds with lung cancer, some early work with a novel nitroimi-
strong electron affinity that mimic the radiosensitizing dazole compound called doranidazole (PR-350) has
effect of oxygen by virtue of their ability to ‘‘fix’’ shown promise in both NSCLC and SCLC. Preclini-
damage produced by free radicals. As with oxygen, cal and early clinical trials have demonstrated a rel-
their strong electron affinity can fix radiation damage. atively high SER in NSCLC, and a recent Japanese
Analogous to the aforementioned OER metric, the phase I/II trial in patients with locally advanced
efficacy of a hypoxic-cell radiosensitizer is expressed NSCLC demonstrated promising efficacy. Patients
numerically as a sensitizer enhancement ratio (SER). receiving 21–30 doses of doranidzole over the course
Misonidazole, a nitroimidazole compound was of the 30 fractions had a median survival of
found to be a potent radiosensitizer of cells in culture, 20.9 months and a 2-year survival rate of 33%
activity which was subsequently confirmed in animal (Nishimura et al. 2007). Furthermore, PR-350 has
studies. However, in the clinical setting, it caused also been shown to effectively radiosensitize SCLC
severe, dose-limiting peripheral neuropathy. Newer in vivo, and further studies are awaited.
generation nitroimidazole compounds such as etani- Modeling parameters suggest that hypoxic cell
dazole and pimonidazole were subsequently devel- radiosensitizers would be of particular benefit in
oped to improve the pharmacokinetic profile of this patients being treated with stereotactic body radiation
class of drugs and reduce toxicity. Unfortunately, therapy (SBRT) (Brown et al. 2010) since the delivery
Radiation Sensitizers 217

of large fractions of radiation over a very short period gliomas and head and neck cancers, where the ability
of time may not allow for sufficient tumor re-oxy- to avoid immediately adjacent critical structures is of
genation. This hypothesis however has not yet been particular importance. Although its use in the treat-
formally tested. ment of lung cancer has not yet been well-studied,
early animal models have confirmed that metastatic
lung lesions have increased uptake of boronated
3.2 Bioreductive Drugs (Hypoxic Cell compounds in comparison to healthy lung tissue,
Cytotoxins) suggesting a possible therapeutic role in this setting
(Bortolussi et al. 2011).
Bioreductive drugs are agents that undergo metabolic
reduction to generate cytotoxic radical species. This
process is facilitated by bioreductive enzymes and the 5 DNA Repair Inhibitors
lower oxygen conditions present in tumors compared
to normal tissues. While it should be mentioned that Radiation therapy functions through its ability to
bioreductive drugs are not radiosensitizers per sé, they damage DNA, directly or indirectly. One mechanism
do however act in a complementary fashion with of therapeutic resistance therefore is the ability of
radiation by selectively targeting and killing hypoxic cancer cells to identify and repair DNA damage. The
cells that would otherwise be radioresistant. The most use of pharmacological agents that can inhibit DNA
well-studied of these compounds is tirapazamine, and repair pathways in cancer cells has the potential to
early lab studies suggested a supra-additive effect counter the phenomenon of therapeutic resistance,
when combined with ionizing radiation (Wilson et al. thus enhancing the cytotoxicity of radiation. Impor-
1996). Unfortunately, efforts to use this compound in tant DNA repair pathways including homologous
clinical trials have been hampered by unpleasant side recombination repair (HRR), non-homologous end-
effects associated with this agent such as nausea and joining (NHEJ), base-excision repair (BER), nucleo-
muscle cramping. Furthermore, the recently com- tide-excision repair (NER), mismatch repair (MMR)
pleted phase III TROG 02.02/HeadSTART trial and translesion DNA synthesis (TLS) (Helleday et al.
adding tirapazamine to chemoradiation in locally 2008). The development of novel agents that can
advanced head and neck carcinoma did not demon- inhibit or modulate these pathways is an area of
strate improved overall survival (Rischin et al. 2010). intense ongoing study. Here, we outline some of the
Tirapazamine has also been evaluated in a recently major classes of drugs that modulate DNA repair.
completed phase II SWOG study for patients with
limited stage SCLC. A median survival of 21 months
was observed in this study (Le et al. 2009). As similar 5.1 AKT Pathway Inhibition
survival rates have been observed with chemoradio-
therapy alone (Turrisi et al. 1999), it is not clear Non-small-cell lung cancers often display amplifica-
whether this compound has merit in SCLC. tion of EGFR (Ohsaki et al. 2000) which stimulates
the phosphatidylinositol 3-kinase (PI3K)/AKT path-
way as well as the Ras-Raf-Mek-ERK signaling cas-
4 Boron Neutron Capture Theory cade, both of which induce radioresistance through
the promotion of DNA repair. For this reason, agents
Boron neutron capture theory (BNCT) involves the such as EGFR-inhibitors (Kriegs et al. 2010; Tanaka
injection of a boron-10 tagged chemical (one that et al. 2008) or AKT-inhibitors (Toulany et al. 2008a)
preferentially binds to tumor cells) followed by irra- have the potential to interfere with DNA repair, and
diation of the tumor target with a neutron beam. The thus may enhance radiation sensitivity through this
interaction of the neutrons with the boron nuclei mechanism.
produces densely ionizing radiation within the cell Interestingly, statins, a commonly prescribed class
(secondary to production of (Nagata et al. 2010) Li of medications used primarily as anti-cholesterol
and an alpha particle). This modality has primarily agents, have also been shown to interfere with AKT
been investigated in the treatment of malignant signaling. In addition to inhibiting cholesterol
218 A. M. Brade and Z. Allibhai

biosynthesis, they might also have chemopreventative gefitinib, again in human NSCLC lines (Tanaka
and cytostatic properties. For example, simvastatin et al. 2008).
has been shown to inhibit growth in both NSCLC
(Bellini et al. 2003) and SCLC (Khanzada et al.
2006). In laboratory studies they have also demon- 5.3 HDAC-Inhibitors
strated the ability to enhance radiosensitivity in cer-
vical cancer and more recently NSCLC. These agents Histone deacetylases (HDAC) modulate the acetyla-
drive apoptosis following exposure to ionizing radi- tion status of histones, thus affecting the gene tran-
ation by simultaneously inhibiting and activating the scription. HDAC overactivity has been associated with
Akt- and AMPK-signaling pathways respectively. tumorigenesis, presumably through transcriptional
In comparison to control cells exposed to either 2 or repression of tumor suppressor genes (Zhang et al.
8 Gy, the use of lovastatin significantly reduced 2004). HDAC-Inhibitors (HDACIs) have been shown
proliferation of NSCLC by 63 and 90% respectively to suppress the ability of cancer cells to repair DSBs in
(Sanli et al. 2011). Direct inhibition of AKT1 has response to ionizing radiation (Marks et al. 2000).
been shown to downregulate radiation-induced DNA- Laboratory studies evaluating different HDACIs
PKcs activity suggesting a potential link between have demonstrated that co-treatment with these agents
statins and DNA DSB repair (Toulany et al. 2008a). increases the radiosensitivity in NSCLC cells (Zhang
et al. 2009; Geng et al. 2006; Cuneo et al. 2007).
Vorinostat, a first generation HCADi does not appear
5.2 Mitogen-Activated Protein Kinase to have single agent activity in lung cancer suggesting
Inhibition that HDACIs may function as true radiosenstizers
(Vansteenkiste et al. 2008). Ongoing and future
The mitogen-activated protein (MAP) kinase cascade studies will help to determine whether this promising
is involved in cancer cell proliferation, survival, and strategy can provide meaningful clinical benefit.
metastases, thus playing an important role in the Histone acetyltransferase (HAT) works in con-
progression and maintenance of cancer (Shannon junction with HDAC to regulate transcription.
et al. 2009). This pathway (which is often upregu- Interestingly, recent studies have shown that HAT-
lated in tumors), is activated following exposure to inhibitors may also have potent radiosensitizing
ionizing radiation, and can modulate DNA-DSB effects (Bandyopadhyay et al. 2009), and further
repair, through its role in the upregulation of X-ray studies are ongoing.
repair cross-complementing group I protein PARP-Inhibitors Poly(ADP-ribose) polymerase
(XRCC1) expression (Toulany et al. 2008b; Wood (PARP) is a nuclear enzyme that plays an important
et al. 2009). Inhibition of the MAP kinase pathway role in sensing and repairing DNA damage via the
has been shown to enhance the radiosensitivity of modification of a number of key proteins, particularly
non-small-cell lung cancer cells. AZD6244 is one those involved in single-strand break repair via BER
such MAP-kinase inhibitor, and an in vivo study of (de Murcia et al. 1997; Dantzer et al. 2000). With
this agent in NSCLC demonstrated an impressive continuous PARP inhibition, single-strand breaks are
SER of 3.38 (Chung et al. 2009). Interestingly, the converted to double-strand breaks, and thus PARP-
radiosensitization observed using EGFR inhibitors inhibitors demonstrate synthetic lethality in tumors
may stem, at least in part from inhibition of DNA- that are reliant on NHEJ and BER (such as those
DSB repair mediated by MAP kinase. Both erlotinib affected by BRCA-1 or BRCA-2 mutations) (Chalmers
and cetuximab have been shown to downregulate et al. 2010). Various PARP inhibitors have been shown
NHEJ repair of DSBs with erlotinib interfering with to potentiate the effect of radiation, particularly in
signaling through both AKT and MAPK but cetux- hypoxic cells whose radiosensitivity is enhanced to a
imab functioning through MAPK in human NSCLC similar degree as oxic cells (Liu et al. 2008). Preclin-
cell lines (Kriegs et al. 2010). This confirmed a ical studies of PARP inhibitors in lung cancer models
similar finding reported previously by Tanaka using have been encouraging (Albert et al. 2007).
Radiation Sensitizers 219

(Cerchietti et al. 2005; Klenke et al. 2011) NSCLC


6 Apoptosis Modulating Agents lesions. These led to the development of a random-
ized phase II trial comparing RT +/- concurrent
6.1 Bcl-2 Inhibitors celecoxib in patients with stages II–III disease (De
Ruysscher et al. 2007). Unfortunately, this study
The Bcl-2 gene family of proteins is fundamentally closed due to poor accrual, and thus it remains unclear
involved in regulation of the apoptotic pathway with as to whether or not this strategy has merit in the
some family members effecting pro-apoptotic sig- clinical setting.
naling and others functioning as anti-apoptotic mod- In conclusion, to date, no clear benefit has been
ulators. While the relative importance of apoptosis (as established for pure radiosensitization strategies in
opposed to mitotic catastrophe) in radiation-induced lung cancer. Many novel approaches in this field have
cytotoxicity in solid tumors is debatable, there is shown recent promise, however. The ability to
some evidence that drugs modulating the apoptotic selectively enhance the radiosensitivity of tumor cells
response may be relevant to radiosensitivity in lung continues to hold appeal, particularly when combined
cancer. For example, radioresistant cells have been with recent technological advances that enable
demonstrated to have higher levels of Bcl-2 (Roa delivery of radiation with increased accuracy and
et al. 2005). Preclinical studies have suggested that precision. Along with improvements in cytotoxic
certain Bcl-2 inhibitors may enhance radiosensitivity chemotherapy and targeted agents, this field remains a
in both NSCLC (Moretti et al. 2010) and SCLC (Hann potentially fruitful area for research in the quest to
et al. 2008; Loriot et al. 2010). In the case of NSCLC, further increase the therapeutic ratio and improve
both radiosensitive and radioresistant cell lines had outcome in patients with lung cancer.
increased response to RT when given with one of
these agents.
References

6.2 Cyclooxygenase COX-2-Inhibitors Albert JM, Cao C, Kim KW et al (2007) Inhibition of


Poly(ADP-Ribose) polymerase enhances cell death and
improves tumor growth delay in irradiated lung cancer
COX-2 mediates prostaglandin synthesis in response models. Clin Cancer Res 13:3033–3042
to various stimuli such as inflammation and exposure Ang KK (2010) More lessons learned from the suffocation of
to carcinogens. This enzyme has been found to be hypoxia. J Clin Oncol 28(18):2941–2943
Bandyopadhyay K, Banères JL, Martin A et al (2009)
upregulated in many tumors including cancer of the Spermidinyl-CoA-based HAT inhibitors block DNA repair
lung (Komaki et al. 2004) where it serves to inhibit and provide cancer-specific chemo- and radio-sensitization.
apoptosis and modulate cell proliferation (Sobolewski Cell Cycle 8(17):2779–2788
et al. 2010). Several preclinical models and Phase I Becker A, Hansgen G, Bloching M et al (1998) Oxygenation of
squamous cell carcinoma of the head and neck: comparison
studies have demonstrated that COX-2 inhibitors of primary tumors, neck node metastases, and normal tissue.
and other non-steroidal anti-inflammatory drugs Int J Radiat Oncol Biol Phys 42:35–41
(NSAIDs) increase radiosensitivity of tumors without Bellini MJ, Polo MP, de Alaniz MJ et al (2003) Effect of
significantly increasing toxicity. For example, ni- simvastatin on the uptake and metabolic conversion of
palmitic, dihomo-gamma-linoleic and alpha-linolenic acids
mesulide (Kim et al. 2009; Grimes et al. 2006) has in A549 cells. Prostaglandins Leukot Essent Fatty Acids
been studied in xenograft models, demonstrating 69:351–357
significantly delayed tumor growth compared to Bennett MH, Feldmeier J, Hampson N, Smee R, Milross C.
tumors receiving radiation alone. The in vitro com- Hyperbaric oxygen therapy for late radiation tissue injury.
The Cochrane Database of Systematic Rev 2005, Issue 3.
ponent of the study showed significantly decreased Art. No.: CD005005.pub2. DOI: 10.1002/14651858.CD0
clonogenic survival with the addition of nimesulide 05005.pub2
(P \ 0.001). Bernier J, Denekamp J, Rojas A et al (1999) ARCON:
Celecoxib is another COX-2 inhibitor studied for accelerated radiotherapy with carboge n and nicotinamide
in non small cell lung cancer: a phase I/II study by the
its radiosensitizing properties. This agent had shown EORTC. Radiother Oncol 52(2):149–156
promise in preclinical and early clinical studies for Bohlius J, Schmidlin K, Brillant C et al (2009) Recombinant
both primary (Liu et al. 2003) and metastatic human erythropoiesis-stimulating agents and mortality in
220 A. M. Brade and Z. Allibhai

patients with cancer: a meta-analysis of randomised trials. Eschmann SM, Paulsen F, Reimold M (2005) Prognostic
Lancet 373(9674):1532–1542 impact of hypoxia imaging with 18F-misonidazole PET in
Bortolussi S, Bakeine JG, Ballarini F et al (2011) Boron uptake non-small cell lung cancer and head- and neck-cancer
measurements in a rat model for boron neutron capture before radiotherapy. J Nucl Med 46(2):253–260
therapy of lung tumors. Appl Radiat Isot 69(2):394–398 Eschwège F, Sancho-Garnier H, Chassagne D et al (1997)
Brizel DM, Rosner GL, Prosnitz LR et al (1995) Patterns and Results of a European randomized trial of Etanidazole
variability of tumor oxygenation in human soft tissue combined with radiotherapy in head- and neck-carcinomas.
sarcomas, cervical carcinomas, and lymph node metastases. Int J Radiat Oncol Biol Phys 39(2):275–281
Int J Radiat Oncol Biol Phys 32:1121–1125 Fyles AW, Milosevic M, Wong R et al (1998) Oxygenation
Brizel DM, Scully SP, Harrelson JM et al (1996) Tumor predicts radiation response and survival in patients with
oxygenation predicts for the likelihood of distant metas- cervix cancer. Radiother Oncol 48:149–156
tases in human soft tissue sarcoma. Cancer Res 56(5): Geng L, Cuneo KC, Fu A et al (2006) Histone deacetylase
941–943 (HDAC) inhibitor LBH589 increases duration of gamma-
Brown JM, Gaccia AJ (1998) The unique physiology of solid H2AX foci and confines HDAC4 to the cytoplasm in
tumors: opportunities (and problems) for cancer therapy. irradiated non-small cell lung cancer. Cancer Res
Cancer Res 58:1408–1416 66(23):11298–11304
Brown JM, Diehn M, Loo BW (2010) Stereotactic ablative Grimes KR, Warren GW, Fang F et al (2006) Cyclooxygenase-
radiotherapy should be combined with a hypoxic cell 2 inhibitor, nimesulide, improves radiation treatment
radiosensitizer. IJROBP 78(2):323–327 against non-small cell lung cancer both in vitro and
Bui QC, Lieber M, Withers HR et al (2004) The efficacy of in vivo. Oncol Rep 16(4):771–776
hyperbaric oxygen therapy in the treatment of radiation- Hann CL, Daniel VC, Sugar EA et al (2008) Therapeutic
induced late side effects. Int J Radiat Oncol Biol Phys efficacy of ABT-737, a selective inhibitor of BCL-2, in
60(3):871–878 small cell lung cancer. Cancer Res 68:2321–2328
Cerchietti LC, Bonomi MR, Navigante AH et al (2005) Phase I/ Helleday T, Petermann E, Lundin C et al (2008) DNA repair
II study of selective cyclooxygenase-2 inhibitor celecoxib pathways as targets for cancer therapy. Nat Rev Cancer
as a radiation sensitizer in patients with unresectable brain 8:193–204
metastases. J Neurooncol 71(1):73–81 Henk JM, Kunkler PB, Smith CW (1977) Radiotherapy and
Chalmers AJ, Lakshman M, Chan N et al (2010) Poly(ADP- hyperbaric oxygen in head- and neck-cancer. Final report of
ribose) polymerase inhibition as a model for synthetic first controlled clinical trial. Lancet 2:101–103
lethality in developing radiation oncology targets. Semin Henke M, Laszig R, Rube C et al (2003) Erythropoietin to treat
Radiat Oncol 20(4):274–281 head- and neck-cancer patients with anaemia undergoing
Chinnaiyan P, Huang S, Vallabhaneni G et al (2005) Mech- radiotherapy: Randomised, double-blind, placebo-con-
anisms of enhanced radiation response following epidermal trolled trial. Lancet 362:1255–1260
growth factor receptor signaling inhibition by erlotinib Hockel M, Knoop C, Schlenger K et al (1993) Intratumoral PO2
(Tarceva). Cancer Res 65(8):3328–3335 predicts survival in advanced cancer of the uterine cervix.
Choy H, Nabid A, Stea B et al (2005) Phase II multicenter Radiother Oncol 26:45–50
study of induction chemotherapy followed by concurrent Hoskin PJ, Rojas AM, Bentzen SM et al (2010) Radiotherapy
efaproxiral (RSR13) and thoracic radiotherapy for patients with concurrent carbogen and nicotinamide in bladder
with locally advanced non-small-cell lung cancer. JCO carcinoma. J Clin Oncol 28(33):4912–4918
23(25):5918–5928 Janssens GO, Terhaard CH, Doornaert PA et al (2011)Acute
Chung EJ, Brown AP, Asano H et al (2009) In vitro and in vivo toxicity profile and compliance to accelerated radiotherapy
radiosensitization with AZD6244 (ARRY-142886), an plus carbogen and nicotinamide for clinical stage T2-4
inhibitor of mitogen-activated protein kinase/extracellular laryngeal cancer: results of a phase III randomized trial. Int
signal-regulated kinase 1/2 kinase. Clin Cancer Res J Radiat Oncol Biol Phys. [Epub ahead of print]
15(9):3050–3057 Kaanders JH, Pop LA, Marres HA et al (1998) Accelerated
Cuneo KC, Fu A, Osusky K, Huamani J, Hallahan DE, Geng L radiotherapy with carbogen and nicotinamide (ARCON) for
(2007) Histone deacetylase inhibitor NVP-LAQ824 sensi- laryngeal cancer. Radiother Oncol 48(2):115–122
tizes human nonsmall cell lung cancer to the cytotoxic Kaanders JH, Bussink J, van der Kogel AJ (2004) Clinical
effects of ionizing radiation. Anticancer Drugs 18:793–800 studies of hypoxia modification in radiotherapy. Semin
Dantzer F, de La Rubia G, Menissier-De Murcia J et al (2000) Radiat Oncol 14:233–240
Base excision repair is impaired in mammalian cells lacking Khanzada UK, Pardo OE, Meier C et al (2006) Potent
Poly(ADP-ribose) polymerase-1. Biochemistry 39: inhibition of small-cell lung cancer cell growth by simva-
7559–7569 statin reveals selective functions of Ras isoforms in growth
de Murcia JM, Niedergang C, Trucco C et al (1997) Require- factor signalling. Oncogene 25:877–887
ment of poly(ADP-ribose) polymerase in recovery from Kim BM, Won J, Maeng KA et al (2009) Nimesulide, a
DNA damage in mice and in cells. Proc Natl Acad Sci USA selective COX-2 inhibitor, acts synergistically with ionizing
94:7303–7307 radiation against A549 human lung cancer cells through the
de Ruysscher D, Bussink J, Rodrigus P et al (2007) Concurrent activation of caspase-8 and caspase-3. Int J Oncol
celecoxib versus placebo in patients with stage II–III non- 34(5):1467–1473
small cell lung cancer: a randomised phase II trial. Klenke FM, Abdollahi A, Bischof M et al (2011) Celecoxib
Radiother Oncol 84(1):23–25 enhances radiation response of secondary bone tumors of a
Radiation Sensitizers 221

human non-small cell lung cancer via antiangiogenesis in Nagata Y, Takahashi A, Ohnishi K et al (2010) Effect of
vivo. Strahlenther Onkol 187(1):45–51 Epub 2010 Dec 23 rapamycin, an mTOR inhibitor, on radiation sensitivity of
Komaki R, Liao Z, Milas L (2004) Improvement strategies for lung cancer cells having different p53 gene status. Int J
molecular targeting: cyclooxygenase-2 inhibitors as radio- Oncol 37(4):1001–1010
sensitizers for non-small cell lung cancer. Semin Oncol 31(1 Nishimura Y, Nakagawa K, Takeda K et al (2007) Phase I/II
Suppl 1):47–53 Trial of sequential chemoradiotherapy using a novel hyp-
Kriegs M, Kasten-Pisula U, Rieckmann T et al (2010) The oxic cell radiosensitizer, Doranidazole (PR-350), in patients
epidermal growth factor receptor modulates DNA double- with locally advanced non–small-cell lung cancer (WJTOG-
strand break repair by regulating non-homologous end- 0002). IJROBP 69(3):786–792
joining. DNA Repair 9(8):889–897 Ohsaki Y, Tanno S, Fujita Y et al (2000) Epidermal growth
Laurie SA, Jeyabalan N, Nicholas G et al (2006) Association factor receptor expression correlates with poor prognosis in
between anemia arising during therapy and outcomes of non-small cell lung cancer patients with p53 overexpres-
chemoradiation for limited small-cell lung cancer. J Thorac sion. Oncol Rep 7:603–607
Oncol 1(2):146–151 Overgaard J (1994) Clinical evaluation of nitroimidazoles as
Le Q, Erler JT, Giaccia A (2007) Tumor hypoxia and modifiers of hypoxia in solid tumors. Oncol Res 6:509–518
metastasis in non-small-cell lung cancers. JTO 2(8):S154– Overgaard J, Hansen HS, Overgaard M et al (1998) A
S155 randomized double-blind Phase III study of nimorazole as
Le QT, Moon J, Redman M, Williamson SK, Lara PN Jr, a hypoxic radiosensitizer of primary radiotherapy in supra-
Goldberg Z, Gaspar LE, Crowley JJ, Moore DF Jr, Gandara glottic larynx and pharynx carcinoma. Results of the Danish
DR (2009) Phase II study of tirapazamine, cisplatin, and head and neck cancer study (DAHANCA) protocol 5–85.
etoposide and concurrent thoracic radiotherapy for limited- Radiother Oncol 46:135–146
stage small-cell lung cancer: SWOG 0222. J Clin Oncol Park SY, Kim YM, Pyo H (2010) Gefitinib radiosensitizes non-
27(18):3014–3019 small cell lung cancer cells through inhibition of ataxia
Lee DJ, Cosmatos D, Marcial VA et al (1995) Results of an telangiectasia mutated. Mol Cancer 9:222
RTOG phase III trial (RTOG 85-27) comparing radiother- Rischin D, Peters LJ, O’Sullivan B et al (2010) Tirapazamine,
apy plus etanidazole with radiotherapy alone for locally cisplatin, and radiation versus cisplatin and radiation for
advanced head- and neck-carcinomas. Int J Radiat Oncol advanced squamous cell carcinoma of the head and neck
Biol Phys 32(3):567–576 (TROG 02.02, HeadSTART): a phase III trial of the Trans-
Leyland-Jones B (2003) BEST Investigators and Study Group, Tasman Radiation Oncology Group. J Clin Oncol
Breast cancer trial with erythropoietin terminated unexpect- 28(18):2989–2995
edly. Lancet Oncol 4:459–460 Roa W, Chen H, Alexander A et al (2005) Enhancement of
Liu W, Chen Y, Wang W et al (2003) Combination of radiation radiation sensitivity with BH3I-1 in non-small cell lung
and celebrex (celecoxib) reducemammary and lung tumor cancer. Clin Invest Med 28(2):55–63
growth. Am J Clin Oncol 26(4):S103–S109 Robnett TJ, Machtay M, Hahn SM et al (2002) Pathological
Liu SK, Coackley C, Krause M et al (2008) A novel poly(ADP- response to preoperative chemoradiation worsens with
ribose) polymerase inhibitor, ABT-888, radiosensitizes anemia in non-small cell lung cancer patients. Cancer J
malignant human cell lines under hypoxia. Radiother Oncol 8:263–267
88(2):258–268 Rubin P, Hanley J, Keys HM et al (1979) Carbogen breathing
Loriot Y, Mordant P, Brown BD et al (2010) Inhibition of BCL- during radiation therapy. The RTOG study. IJROBP
2 in small cell lung cancer cell lines with oblimersen, an 5:1963–1970
antisense BCL-2 oligodeoxynucleotide (ODN): in vitro and Sanli T, Liu C, Rashid A et al (2011) Lovastatin sensitizes lung
in vivo enhancement of radiation response. Anticancer Res cancer cells to ionizing radiation: modulation of molecular
30(10):3869–3878 pathways of radioresistance and tumor suppression. J Thorac
MacRae R, Shyr Y, Johnson D (2002) Declining hemoglobin Oncol 6(3):439–450
during chemoradiotherapy for locally advanced non-small Scott C, Suh J, Stea B et al (2007) Improved survival, quality of
cell lung cancer is significant. Radiother Oncol 64(1):37–4 life, and quality-adjusted survival in breast cancer patients
Manegold C, von Pawel J, Zaitloukal P et al (2008) B017704: a treated with efaproxiral (Efaproxyn) plus whole-brain
phase III randomized study of first-line bevacizumab radiation therapy for brain metastases. AJCO 30(6):580–587
combined with cisplatin/gemcitabine (CG) in patients (pts) Shannon AM, Telfer BA, Smith PD et al (2009) The mitogen-
with advanced or recurrent non-squamous, non-small cell activated protein/extracellular signal-regulated kinase
lung cancer (NSCLC). Ann Oncol 19:viii1 kinase 1/2 inhibitor AZD6244 (ARRY-142886) enhances
Marks PA, Richon VM, Rifkind RA et al (2000) Histone the radiation responsiveness of lung and colorectal tumor
deacetylase inhibitors: inducers of differentiation or apop- xenografts. Clin Cancer Res 15:6619–6629
tosis of transformed cells. JNCI J Natl Cancer Inst Sobolewski C, Cerella C, Dicato M et al (2010) The role of
92(15):1210–1216 cyclooxygenase-2 in cell proliferation and cell death in
Moretti L, Li B, Kim KW et al (2010) AT-101, a pan-Bcl-2 human malignancies. Int J Cell Biol 2010:215158
inhibitor, leads to radiosensitization of non-small cell lung Suh JH, Stea B, Nabid A et al (2006) Phase III study of
cancer. J Thorac Oncol 5(5):680–687 efaproxiral as an adjunct to whole-brain radiation therapy
Mok TS, Wu YL, Thongprasert S et al (2009) Gefitinib or for brain metastases. JCO 24(1):106–114
carboplatin paclitaxel in pulmonary adenocarcinoma. Tanaka T, Munshi A, Brooks C et al (2008) Gefitinib
N Engl J Med 361:947–957 radiosensitizes non-small cell lung cancer cells by
222 A. M. Brade and Z. Allibhai

suppressing cellular DNA repair capacity. Clin Cancer Res Watson ER, Halnan KE, Dische S et al (1978) Hyperbaric
14(4):1266–1273 oxygen and radiotherapy: a medical research council trial in
Toulany M, Kehlbach R, Florczak U et al (2008a) Targeting of carcinoma of the cervix. Br J Radiol 51:879–887
AKT1 enhances radiation toxicity of human tumor cells by Wilson WR, Denny WA, Pullen SM et al (1996) Tertiary amine
inhibiting DNA-PKcs-dependent DNA double-strand break N-oxides as bioreductive drugs: DACA N-oxide, nitracrine
repair. Mol Cancer Ther 7(7):1772–1781 N-oxide and AQ4 N. Br J Cancer (Suppl 27):S43–S47
Toulany M, Dittmann K, Fehrenbacher B et al (2008b) PI3K- Wood CD, Thornton TM, Sabio G et al (2009) Nuclear
Akt signaling regulates basal, but MAP-kinase signaling localization of p38 MAPK in response to DNA damage. Int
regulates radiation-induced XRCC1 expression in human J Biol Sci 5(5):428–437
tumor cells in vitro. DNA Repair 7(10):1746–1756 Yamamoto M, Izumi Y, Horinouchi H et al (2009) Systemic
Tsao MS, Sakurada A, Cutz JC et al (2005) Erlotinib in lung administration of hemoglobin vesicle elevates tumor tissue
cancer—molecular and clinical predictors of outcome. oxygen tension and modifies tumor response to irradiation.
N Engl J Med 353(2):133–144 J Surg Res 151(1):48–54
Turrisi AT, Kim K, Blum R et al (1999) Twice-daily compared Zhang Y, Adachi M, Zhao X et al (2004) Histone deacetylase
with once-daily thoracic radiotherapy in limited small-cell inhibitors FK228, N-(2-aminophenyl)-4-[N-(pyridin-3-yl-
lung cancer treated concurrently with cisplatin and etopo- methoxycarbonyl)amino- methyl] benzamide and m-carb-
side. N Engl J Med 340(4):265–271 oxycinnamic acid bis-hydroxamide augment radiation-
Vansteenkiste J, Pirker R, Massuti B et al (2002) Double-blind, induced cell death in gastrointestinal adenocarcinoma cells.
placebo-controlled, ran-domized phase III trial of darbe- Int J Cancer 110(2):301–308
poetin-alfa in lung cancer patients receiving chemotherapy. Zhang F, Zhang T, Teng ZH et al (2009) Sensitization to
JNCI 94:1211–1220 gamma-irradiation-induced cell cycle arrest and apoptosis
Vansteenkiste J, Van Cutsem E, Dumez H et al (2008) Early by the histone deacetylase inhibitor trichostatin a in non-
phase II trial of oral vorinostat in relapsed or refractory small cell lung cancer (NSCLC) cells. Cancer Biol Ther
breast, colorectal, or non-small cell lung cancer. Invest New 8(9):823–831
Drugs 26(5):483–488
Radioprotectors and Chemoprotectors
in the Management of Lung Cancer
Ritsuko Komaki, Zhongxing Liao, James D. Cox,
Kathy A. Mason, and Luka Milas

Contents 12 Concluding Remarks ............................................... 238


References.......................................................................... 239
1 Introduction.............................................................. 224
2 Thiols as Radioprotective Agents .......................... 225
Abstract
2.1 Amifostine: Preclinical Findings............................... 226
2.2 Amifostine: Clinical Studies ..................................... 227 Lung cancer is the leading cause of cancer death in
2.3 Amifostine: Meta-Analyses of Clinical Studies ....... 229 most developed countries. The prognosis remains
2.4 Amifostine: Route of Administration ....................... 230 poor with an overall survival rate at 5 years of only
2.5 Amifostine: Quality of Life Studies ......................... 231
about 15%. Between 70 and 85% of all cases are
3 Prostanoids, COX-2 and COX-2 Inhibitors ......... 231 histologically classified as non-small cell lung
3.1 Clinical Trials of COX-2 Inhibitors carcinoma (NSCLC). Radiation therapy has tradi-
for Lung Cancer ........................................................ 232
tionally been the treatment of choice for locally
4 Growth Factors and Cytokines.............................. 233 advanced disease and medically inoperable early
4.1 Basic Fibroblast Growth Factor ................................ 233
4.2 Keratinocyte Growth Factor...................................... 233
stage NSCLC. However radiation therapy alone
4.3 Interleukin-11............................................................. 234 was not effective treatment for patients with
4.4 Epidermal Growth Factor Receptor.......................... 234 locally advanced NSCLC. The addition of cyto-
5 Pentoxifylline ............................................................ 234 toxic drugs to radiotherapy considerably improves
treatment outcome, and the combination of
6 Angiotensin-Converting Enzyme Inhibitors ......... 235
chemotherapy with radiotherapy has become com-
7 Flavopiridol .............................................................. 236 mon practice for the treatment of locally advanced
8 Poly(ADP-Ribose) Polymerase Inhibitors............. 236 lung cancer. The addition of chemotherapy to
9 Bcl-2 Inhibitors ........................................................ 237
radiotherapy has two principal objectives: one, to
increase the chance of local tumor control and two,
10 Efaproxaril................................................................ 237 to eliminate metastatic disease outside of the
11 Radioprotective Gene Therapy/Antioxidant radiation field. Several randomized trials have
Therapy: Superoxide Dismutase............................ 238 shown improvement of local control and survival
by application of concurrent chemotherapy rather
than sequential chemotherapy followed by radia-
R. Komaki (&)  Z. Liao  J. D. Cox
Department of Radiation Oncology, Unit 97, tion treatment. This combined treatment approach
The University of Texas MD Anderson Cancer Center, results in median survival times of 13 to 14 months
1515 Holcombe Blvd, Houston, TX 77030-4009, USA and survival rates at 5 years as high as 15 to 20%.
e-mail: rkomaki@mdanderson.org These improvements have been achieved by using
K. A. Mason  L. Milas standard chemotherapeutic agents, primarily cis-
Department of Experimental Radiation Oncology, platin-based drug combinations. However, concur-
The University of Texas MD Anderson Cancer Center,
Houston, TX, USA rent chemoradiotherapy has increased significant

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 223
DOI: 10.1007/174_2011_288, Ó Springer-Verlag Berlin Heidelberg 2011
224 R. Komaki et al.

normal tissue toxicity such as esophagitis and achieved by reducing cell burden in tumors under-
pneumonitis. Therefore normal tissue protectors going radiotherapy or by interfering with tumor cell
without protective cancer cells became necessary to radioresistance factors, thereby rendering tumor cells
improve therapeutic ratio. We will discuss mecha- more susceptible to destruction by radiation. Factors
nism and efficacy of Amifostine to protect normal that contribute to tumor radioresistance include the
tissue followed by other normal tissue protectors or failure of tumor cells to undergo cell death after
molecular targeted treatment without increasing radiation, the cells’ ability to efficiently repair DNA
normal tissue damage e.g., prostanoids (COX-2) damage, continued cell proliferation during the course
inhibitors, Growth factor and Cytokines inhibitors, of radiotherapy, cell radioresistance secondary to the
Basic and other inhibitors targeting Fibroblast hypoxia that commonly develops in solid tumors, and
Growth Factor, Karatinocyte Growth Factors, the presence in tumor cells of various abnormal
Epidermoid Growth Factor Receptor, Pentoxifyl- molecular structures or with dysregulated processes
line, Angiotensin-Converting Enzyme Flavopiridol linked to cellular radioresistance (Milas et al. 2003a).
Poly(ADP-Ribose) Polymerase, Bcl-2 , and Efa- The addition of induction (neoadjuvant) chemo-
proxaril, as well as Radioprotective Gene Therapy/ therapy to radiotherapy results in an increase in
Antioxidant Therapy: and Superoxide Dismutase median survival time of approximately 4 months, and
the overall survival rates at 2 years range from 10 to
15% (Milas et al. 2003a; Dillman et al. 1990;
LeChevalier et al. 1991). These modest therapeutic
gains can be improved upon still further by using
1 Introduction concurrent chemoradiotherapy, i.e., by administering
cytotoxic drugs during the course of radiation treat-
Lung cancer is the leading cause of cancer death in ment (Milas et al. 2003a; Komaki et al. 2002; Turrisi
most developed countries. An estimated 1.6 million et al. 1999; Schaake-Konig et al. 1992; Curran et al.
new cases of lung cancer were diagnosed in 2008, and 2003). This combined treatment approach results in
the prognosis remains poor with an overall survival median survival times of 13 and 14 months and sur-
rate at 5 years of only about 15% (Jemal et al. 2004; vival rates at 5 years as high as 15 to 20%. These
American Cancer Society 2011). Between 70 and improvements have been achieved by using standard
85% of all cases are histologically classified as non- chemotherapeutic agents, primarily cisplatin-based
small cell lung carcinoma (NSCLC), which comprises drug combinations. Since the completion of trials that
squamous cell, adenocarcinoma, large cell, or undif- directly compared induction with concurrent chemo-
ferentiated histologic subtypes; the remainder of the radiotherapy and clearly demonstrated the therapeutic
cases are of small cell histology. At the time of superiority of the latter approach (Schaake-Konig
diagnosis most patients present with locally advanced et al. 1992; Curran et al. 2003), concurrent chemo-
disease and many have overt metastatic dissemina- radiotherapy can be regarded as the current standard
tion. Radiation therapy has traditionally been the of care for local-regionally advanced lung cancer.
treatment of choice for locally advanced disease but Yet, the low overall survival rates among patients
has provided limited benefits both in terms of local with lung cancer necessitates the introduction of
tumor control and patient survival, with 2- to 5-year treatment strategies that would further improve local
survival rates of 10% or less (Dillman et al. 1996). tumor control, survival rates, and quality of life.
The addition of cytotoxic drugs to radiotherapy con- Many factors, known and unknown, limit thera-
siderably improves the treatment outcome, and the peutic success of radiotherapy or chemoradiotherapy
combination of chemotherapy with radiotherapy has for lung cancer, with one major factor being the level
become the common practice for the treatment of of tolerance of normal tissues to the damage by these
advanced lung cancer (O’Rourke et al. 2010). The agents. Toxicities associated with chemotherapy and
addition of chemotherapy to radiotherapy has two radiotherapy can limit the dose and duration of the
principal objectives: one, to increase the chance of treatment, adversely affect both short- and long-term
local tumor control and two, to eliminate metastatic quality of life of the patient, be life-threatening, and
disease outside the radiation field. The former can be increase the costs of patient care. Normal tissue
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 225

Table 1 Radioprotective thiols and phosphorothioates


Compound CAS register number Structure
Thiols
Dithiothreitol (DTT) [27565-41-9] HSCH2CH(OH)CH(OH)CH2SH
2-Mercaptoethanol (WR-15504) [60-24-2] HOCH2CH2SH
Cysteamine (MEA, WR-347) [156-57-0] H2NCH2CH2SH
2-((Aminopropyl)amino)ethanethiol (WR-1065) [31098-42-7] H2N(CH2)3NHCH2CH2SH
WR-255591 [117062-90-5] CH3NH(CH2)3NHCH2CH2SH
WR-151326 [120119-18-8] CH3NH(CH2)3NH(CH2)3SH
Phosphorothioates
WR-638 [3724-89-8] H2NCH2CH2SPO3H2
WR-2721 [20537-88-6] H2N(CH2)3NHCH2CH2SPO3H2
WR-3689 [20751-90-0] CH3NH(CH2)3NHCH2CH2SPO3H2
WR-151327 [82147-31-7] CH3NH(CH2)3NH(CH2)3SPO3H2
WR Walter Reed Army Institute of Research
Reprinted from Murray and McBride (1996) with permission

toxicities are more common and more serious after The first part of the discussion focuses on the radio-
chemoradiotherapy than after radiotherapy alone, and protective effects of amifostine. Additional informa-
these toxicities can be particularly severe with con- tion can be found in other reviews on this topic
current (as opposed to sequential) chemoradiotherapy. (Kouvaris et al. 2007; Weiss and Landauer 2009).
Because of the risk of severe toxicity, the dose of Other radioprotective agents and potential strategies
chemotherapeutic agents in the setting of concurrent discussed include prostanoids, growth factors and
chemoradiotherapy must be significantly reduced, cytokines, pentoxifylline, angiotensin-converting
which may reduce the ability of the drugs to control enzyme inhibitors, and gene therapy focused on
both local-regional tumor and disseminated disease. superoxide dismutase.
Because normal tissue toxicity is a major barrier to
radiotherapy and chemoradiotherapy for lung cancer,
every effort must be taken to avert or minimize 2 Thiols as Radioprotective Agents
potential injury to critical normal tissues or other side
effects of these treatments. Improvements are being Both preclinical and clinical investigations on chem-
sought primarily through more precise delivery of ical protectors in radiotherapy have predominantly
radiation therapy or the use of chemical or bio- focused on thiols. The most effective compounds have
logical radioprotective or chemoprotective agents. those with a sulfhydryl, SH, group at one terminus
Technical improvements in radiotherapy include and a strong basic function, an amino group, at the
three-dimensional treatment planning, conformational other terminus. Some important radioprotective
radiotherapy techniques such as intensity-modulated thiols are listed in Table 1. The general structure of
radiotherapy, or the use of proton beam therapy. these aminothiols is H2N(CH2) x NH(CH2)y SH;
These normal tissue-sparing strategies may allow among these, phosphorothiaotes (such as WR 2721,
higher doses of radiation, chemotherapeutic drugs, or WR-3689, WR-151327) are the most effective and
both to be given, with the hope of achieving superior least toxic (Murray and McBride 1996). Various
treatment outcome. mechanisms have been proposed for the thiol-medi-
This chapter is intended as an overview of selected ated radiation protection of normal tissues. Thiols
relevant preclinical and clinical findings on the use of (RSH) and their anions (RS-) rapidly bind to free
radio- and chemo-protective agents to prevent or radicals such as OH and prevent them from reacting
reduce injury to normal tissues that limit the use with cellular DNA. This type of protection from DNA
of cytotoxic doses of radiotherapy for lung cancer. damage by scavenging free radicals is oxygen-
226 R. Komaki et al.

dependent (Travis 1984). Another mode of protec- 1996; Milas et al. 1988). Amifostine was also a
tion occurs via H-atom donation (the fixation–repair potent radioprotector of late-responding tissues such
model). Thiols compete with oxygen for radiation- as lung and subcutaneous tissues (Milas et al. 1988;
induced DNA radicals. DNA radicals are ‘‘fixed’’ Travis et al. 1985; Vujaskovic et al. 2002a, 2002b;
(not repaired) by reacting with oxygen and poten- Lockhart 1990; Hunter and Milas 1983). Protection
tially harmful hydroxyperoxides may be generated. of the lung was achieved against both single and
However, DNA radicals can be chemically repaired fractionated radiation, and was assessed by bio-
when they react with thiols by donation of hydro- chemical testing such as reduction in hydroxyproline
gen (Durand 1983). Further, intracellular oxygen content of lung tissue and functional assays such as
can be depleted as a result of thiol oxidation breathing frequency (Travis et al. 1985; Vujaskovic
(Durand and Olive 1989), which would decrease the et al. 2002a). Amifostine treatment was associated
rate of oxygen-mediated DNA damage fixation. with reduction in accumulation of macrophages in
Finally, thiols induce DNA packaging that may irradiated lung and profibrogenic cytokine activity
decrease the accessibility of DNA sites to radiolytic (Vujaskovic et al. 2002b). Interestingly, systemic
attack. This mechanism may be oxygen-independent application of amifostine was radioprotective for the
and may explain the protection from densely ion- lung tissue (Travis et al. 1985; Vujaskovic et al.
izing radiation such as neutrons (Savoye et al. 2002a, b), but inhaled amifostine was ineffective
1997). (Lockhart 1990). In contrast to the near- universal
protection of acutely responding tissues and lung,
amifostine was not effective in protecting the brain
2.1 Amifostine: Preclinical Findings from radiation injury, which was attributed to the
inability of the hydrophylic drug to cross the blood-
Amifostine (Ethyol) is a thiol-containing compound brain barrier (Utley et al. 1984). Wide variation in
that has long been recognized for its strong radio- the degree of radioprotection was noted among
protective properties and has been used in clinical various tissues, with protection factors for murine
trials (Brizel 2003, 2007; Movsas et al. 2005). normal tissues ranging from 1.2 for hair follicles to
Amifostine does not readily cross the cell membrane greater than two for jejunum and bone marrow
because of its hydrophilicity. The drug is rapidly (Murray and McBride 1996; Milas et al. 1988). The
dephosphorylated to its active metabolite WR-1065 degree of radioprotection depended on the drug dose
and cleared from plasma with a half-life of 1 to 3 min and time of administration in relation to radiation
after intravenous administration (Shaw et al. 1999). exposure. In general, higher doses of amifostine
In contrast to its brief systemic half-life, the drug is produced better protection up to a maximum dose of
retained for prolonged periods in normal tissues about 400 mg/kg (Murray and McBride 1996; Milas
(Yuhas 1980). During the first 30 min after adminis- et al. 1982, 1988). Maximum radioprotection was
tration, drug uptake into normal tissues such as the achieved when amifostine was given 10 to 30 min
salivary gland, liver, kidney, heart, and bone marrow before radiotherapy (Murray and McBride 1996;
has been demonstrated to be up to 100-fold greater Milas et al. 1988, 1982). In addition to normal tissue
than in tumor tissues (Yuhas 1980). Biodistribution radioprotection, several studies have examined
studies show that the highest tissue levels of amifos- whether amifostine protects tumors as well.
tine and its metabolites are found in salivary glands Although some studies documented a small degree
(Rasey et al. 1986). of tumor radioprotection, primarily of microscopic
During the 1970s and 1980s extensive animal tumor foci, most studies showed no tumor protection
studies explored the ability of amifostine to protect a (Murray and McBride 1996; Milas et al. 1982, 1988;
variety of normal tissues against acute and late Wasserman et al. 1981). Therefore, preclinical
radiation injury and whether the drug improves studies support the notion of selective or preferential
therapeutic ratio of radiotherapy. A radioprotective normal tissue protection resulting in increased ther-
effect was observed for acute injury of the bone apeutic gain of radiotherapy.
marrow, esophagus, jejunum, colon, hair follicles, The mechanism of amifostine’s selective or pref-
testis, and immune system (Murray and McBride erential protection of normal tissues is related to
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 227

several factors. Amifostine undergoes preferential duration of xerostomia in head and neck cancer
rapid uptake into normal tissues but negligible or patients treated with radiotherapy. The findings from
slower uptake into tumor tissues. Whereas normal that study led the US Food and Drug Administration
tissues actively concentrate amifostine against the to approve the use of amifostine for this clinical
concentration gradient, solid tumors generally absorb indication.
amifostine passively (Yuhas 1980). This selectivity Several clinical trials have been performed using
results, in part, from differences in pH and alkaline amifostine in combination with chemoradiotherapy
phosphatase at the level of the capillary endothelium, for lung cancer (Koukourakis et al. 2000; Antonadou
both being higher in normal tissues than in tumors et al. 2001, 2003, Senzer 2002; Leong et al. 2003;
(Yuhas 1980; Rasey et al. 1985, 1986). The acidic Komaki et al. 2004a; Movsas et al. 2005). Results are
tumor microenvironment inhibits the alkaline phos- summarized in Table 2. Antonadou et al. (2001,
phatase necessary for uptake and conversion of ami- 2003) conducted a randomized phase III trial of
fostine to the active protective thiol WR-1065 concurrent chemotherapy (either paclitaxel or carbo-
(Calabro-Jones et al. 1985), a condition absent in platin) and radiation treatment plus/minus daily ami-
normal tissues. Once inside the cell, WR-1065 acts as fostine given iv at 300 mg/m2 15 to 20 min before
a scavenger of oxygen free radicals (Marzatico et al. each fraction of radiotherapy and before chemother-
2000), which is reduced under hypoxic conditions apy in patients with locally advanced lung cancer.
commonly present in solid tumors. In addition, ami- The results showed that amifostine significantly
fostine may be less available to tumors because of reduced the incidence of severe (grade C3) radiation-
their defective vascular network. induced pneumonitis (from 56.3 to 19.4%, P \ 0.002)
Overall, a large body of preclinical data shows that and (grade C3) esophagitis (from 84.4 to 38.9%,
amifostine preferentially protects most normal tissues, P \ 0.001) without compromising antitumor efficacy
including the lung, from the effects of DNA-damag- (Antonadou et al. 2003).
ing agents such as radiation. In addition to inter- Komaki et al. (2004a) reported findings from a
acting with radiation, amifostine has been shown to prospective randomized comparative study of whe-
exert independent antimetastatic and antiangiogenic ther amifostine could reduce the incidence and
activity (Grdina et al. 2002; Giannopoulou and severity of acute toxicity associated with concurrent
Papadimitriou 2003). Thus, these preclinical data cisplatin-based chemotherapy with radiation therapy
provide a strong rationale for the clinical development for NSCLC. In that study, 62 evaluable patients (of 64
of combined modality cancer treatment with amifos- total) received 69.6 Gy of thoracic radiation at
tine and radiotherapy. 1.2 Gy/fraction, 2 fractions/day for 5 days/week;
chemotherapy consisted of oral etoposide, 50 mg
twice a day for a total of 20 days plus four doses of
2.2 Amifostine: Clinical Studies cisplatin 50 mg/m2 iv. The experimental group
received amifostine, 500 mg iv, twice weekly before
Clinical trials with amifostine began in the 1980s and chemoradiation, and the control group received
showed that the drug is generally well tolerated. Its chemoradiation without amifostine. At a minimum
administration is associated with several transient side follow-up time of 24 months (median 31 months for
effects including nausea, vomiting, sneezing, mild living patients), amifostine increased the incidence of
somnolence, hypotension, a metallic taste during mild esophageal toxicity (from 23 to 48%), but con-
infusion, and occasional allergic reactions (Kligerman versely it reduced the incidence of severe esophageal
et al. 1988; Schuchter and Glick 1993). Hypotension toxicity (from 35 to 16%) (P = 0.021) (Fig. 1).
seemed to be the most clinically significant side effect Amifostine significantly reduced the incidence of
that could curtail treatment. Several subsequent trials constipation, pneumonitis, and neutropenic fever
showed that amifostine reduces the severity of (Fig. 2). Notably, severe (grade 3) pneumonitis
toxicity of radiotherapy or chemotherapy (Kligerman occurred in 16% of patients treated with chemora-
et al. 1988; Brizel et al. 2000; Kemp et al. 1996). diotherapy alone but in no patients that received
Brizel et al. (2000) reported a randomized trial amifostine in addition to chemoradiotherapy. Con-
showing that amifostine reduces both severity and sistent with findings from other studies, hypotension
228 R. Komaki et al.

Table 2 Randomized trials of amifostine for lung cancer


Reference No. of Radiation dose Chemotherapy Amifostine dose Comments
patients
Movsas 242 69.6 Gy at Induction P ? C 9 2; 500 mg iv 4 9 per No difference in
et al. 1.2 Gy twice concurrent weekly C week between twice- esophagitis but improved
(2005) daily, days daily RT fractions self-reported swallowing
43–78 and weight loss
Median survival, 15.6
months with versus 17.9
months without amifostine
Antonadou 146 55–60 Gy at None 340 mg/m2 daily ; pneumonitis
et al. 2.0 Gy once before RT ; esophagitis
(2001) daily
(no survival data)
2
Antonadou 73 55–60 Gy at Concurrent weekly P or C 300 mg/m daily ; esophagitis (P \ 0.001)
et al. 2.0 Gy once before chemoRT and ; pneumonitis (P = 0.009)
(2003) daily RT
(no survival data)
Senzer 63 64.8 Gy at Concurrent P ? C q week 500 mg iv before No difference in toxicity,
(2002) 1.8 Gy once 9 7; gemcitabine and weekly chemo; no survival data (ongoing
daily, beginning cisplatin 9 3 after 200 mg iv daily trial)
on day 1 chemoradiation before RT
Komaki 62 69.6 Gy at Concurrent iv cisplatin 500 mg iv day 1 and ; degree of esophagitis
et al. 1.2 Gy twice day 1, 8, 29, 36 2 each week before ; pneumonitis
(2004a) daily, beginning chemo and first RT
on day 1 fraction ; neutropenic fever
Etoposide po days 1–5 Median survival, 19
8–12, 29–33, 36–40 months with versus 20
months without
amifostine)
Leong et al. 60 60–66 Gy at Induction P ? C 9 2; 740 mg/m2 with each Esophagitis grade 2–3
(2003) 2.0 Gy once concurrent weekly P chemo (day 1, 22, 43, 43% in amifostine group,
daily, beginning 50, 57, 64, 71, 78) 70% in control group (not
on day 43 significant)
Median survival, 12.5
months with versus 14.5
months without amifostine
P paclitaxel, C carboplatin; RT radiation therapy
Adapted from Komaki et al. (2004a), with permission

was the most common adverse effect, occurring in (Movsas et al. 2005), 243 patients with stages
65% of patients. Importantly, amifostine did not affect II–IIIA/B NSCLC were treated with paclitaxel
tumor response to chemoradiotherapy, as demon- and carboplatin as induction chemotherapy followed
strated by rates of local-regional control, distant by concurrent chemotherapy and hyperfractionated
metastases-free survival, and overall survival (Fig. 3). radiotherapy (69.6 with 1.2 Gy/fraction, twice a day).
Other investigations from MD Anderson showed that Patients received either no-amifostine or amifostine
amifostine can partially reverse the reduction of lung 500 mg iv four times/week. The amifostine group
diffusion capacity caused by chemotherapy or radio- experienced higher rates of acute nausea, vomiting,
therapy (Gopal et al. 2003), further documenting cardiovascular toxicity, and infection or febrile neu-
amifostine-induced radioprotection of normal tissues tropenia, but interestingly patient self-assessments
during thoracic radiotherapy. indicated that amifostine was associated with clini-
In another important clinical trial, the phase III cally meaningful improvements in swallowing and
Radiation Therapy Oncology Group (RTOG) 98-01 pain. Amifostine did not reduce the rates of severe
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 229

50 to having protective effects on normal tissues, would


Percent Experiencing Toxicity

also have protective effects on tumors, thereby affect-


40
ing the efficacy of therapy. To date, three meta-analyses
have been conducted to address this question. In the
30
Control first of these analyses (Sasse et al. 2006), the investi-
Amifostine gators reviewed 14 randomized trials (1451 patients)
20
that compared radiation with or without amifostine for
10
treatment of a variety of types of cancer. They found
that the use of amifostine significantly reduced the risk
0
of developing mucositis, esophagitis, acute xerosto-
Mild Moderate Severe mia, late xerostomia, dysphagia, acute pneumonitis,
Degree of Esophageal Toxicity and cystitis. Although no difference in the overall
response rate was noted between the amifostine and
Fig. 1 Effect of amifostine on the severity of esophageal
toxicity induced by chemoradiotherapy in patients with non- no-amifostine groups, the complete response rate was
small cell lung cancer (modified from Komaki et al. 2004a, thought to be superior for patients using amifostine.
with permission) The next meta-analysis focused specifically on
patients with locally advanced NSCLC who had been
Percent Experiencing Toxicity

100
treated with radiation or chemoradiation with or
without amifostine or a placebo (Mell et al. 2007).
80
This analysis of six studies (552 patients) revealed
60
Control
that amifostine was associated with slightly improved
40 Amifostine response rates, with pooled relative risk estimates
ranging from 0.99 to 1.21, leading the investigators to
20
conclude that amifostine has no effect on tumor
0 response in locally advanced NSCLC.
Constipation Pneumonitis Neutropenic fever
A third meta-analysis (Bourhis et al. 2011) was
Fig. 2 Effect of amifostine on the incidence of constipation, undertaken with the rationale that neither of the first
pneumonitis, and neutropenic fever caused by chemoradiother- two analyses had access to survival data. This meta-
apy in patients with non-small cell lung cancer (modified from
Komaki et al. 2004a, with permission.) analysis, which involved updating individual patient
data from the surveyed trials, was intended to
evaluate the effect of amifostine on overall and
progression-free survival. Ultimately 12 trials (1,119
esophagitis, but it also did not affect median survival patients) were analyzed; 431 patients (3 trials) were
time. These investigators concluded that other, less treated with radiation only and 688 (9 trials)
toxic routes of administration of amifostine should be received chemoradiation. About one-third of the
explored, as well as other means of minimizing patients had lung cancer; most of the others had
esophagitis such as reducing esophageal exposure head and neck cancer. The vast majority of patients
with more conformal radiation techniques and (91%) had locally advanced disease. The median
exploring various biological therapies. follow-up time was 5.2 years. The results showed
that amifostine did not reduce overall or progression-
free survival in patients treated with radiation or
2.3 Amifostine: Meta-Analyses of Clinical chemoradiation therapy—in other words, that ami-
Studies fostine had no tumor-protective effect. They did note
that any benefit in terms of reduced radiation-
Although the findings from clinical studies of amifos- induced toxicity from amifostine must be weighed
tine are intriguing, the median follow-up times have against the costs and side effects of this agent, par-
been relatively short and none of the randomized con- ticularly given the ongoing improvements in tech-
trolled trials conducted to date has been powered to nologies that allow better sparing of normal tissues
address the question of whether amifostine, in addition (Brizel 2007).
230 R. Komaki et al.

Fig. 3 Kaplan–Meier A. 100


survival curves showing the
75 P=0.99
effect of amifostine on

% Survival
a overall survival rate, 50 Amifostine
b local-regional (LR) tumor
25
control, and c distant- Control
metastasis (DM)-free survival 0
among patients with non- 0 1 2 3
small cell lung cancer after
B. 100
chemoradiotherapy (modified

% LR Control
from Komaki et al. 2004a, 75
P=0.39
with permission) Control
50
25
Amifostine
0
0 1 2 3
C.
% DM-Free Survival

100
P=0.77
75
Control
50
25 Amifostine
0
0 1 2 3

2.4 Amifostine: Route of Administration Another study by the same group showed that sub-
cutaneous amifostine given before chemotherapy
Although most of the larger clinical trials of amifos- was better tolerated than the intravenous form
tine conducted to date have involved intravenous (Koukourakis et al. 2003). A French randomized study
administration, its associated adverse effects (e.g., conducted by the GORTEC group (Bardet et al. 2002)
Movsas et al. 2005) as well as logistic issues have led compared amifostine given in intravenous doses of
to exploration of alternative routes of administration 200 mg/m2 and subcutaneous doses of 500 mg in 305
(Praetorius and Mandal 2008; Bensadoun et al. 2006). patients with head and neck carcinoma. The results of
Of the three routes tested in clinical settings to date this study, completed in December 2004, suggest that
(oral, subcutaneous, and intrarectal), most studies subcutaneous injection is feasible, only mildly toxic,
have focused on subcutaneous delivery. A phase I reduces treatment-related side effects relative to other
study to compare the relative bioavailability of ami- published series, and does not diminish antitumor
fostine administered subcutaneously (at a fixed dose efficacy. A phase II study of the efficacy of subcuta-
of 500 mg) and intravenously (200 mg/m2) (Bonner neous administration of amifostine in patients with
and Shaw 2002) suggested that the bioavailability of surgically resected NSCLC undergoing postoperative
the product is greater when injected subcutaneously radiotherapy is ongoing at MD Anderson. Other
rather than intravenously. In a randomized phase II studies are being conducted at Kaiser Permanente in
study of 140 patients to assess the feasibility, toler- California (Wang et al. 2004) and at Dana Farber in
ance, and cytoprotective efficacy of subcutaneous Boston (Haddad et al. 2003) to evaluate the safety and
amifostine after radiation therapy (Koukourakis et al. efficacy of subcutaneous amifostine during chemora-
2000), 70 patients received 500 mg of amifostine as a diation for head and neck cancer. Finally, a recent
single subcutaneous injection 20 min before each report of a study of patients with small cell lung cancer
radiotherapy fraction. The regimen was well toler- from Korea (Han et al. 2008) suggested that subcuta-
ated, effectively reduced the early toxicity of radio- neous amifostine (500 mg, 3 times a week during
therapy, and prevented treatment-induced delays. concurrent chemoradiation therapy) was tolerated
Patients reported a reduction in hypotension and with manageable adverse effects but did not reduce
nausea as compared with intravenous administration. severe treatment-related toxicities.
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 231

2.5 Amifostine: Quality of Life Studies mononuclear cells such as macrophages and mediates
the typical symptoms of inflammation through its
The 2008 American Society of Clinical Oncology vasodilatory action. This augments the edema caused
practice guidelines (Hensley et al. 2009) recommend by substances that increase vascular permeability
that amifostine not be used to prevent esophagitis such as histamine. PGE2 is also involved in the
during concurrent chemoradiotherapy for NSCLC development of erythema and heat at the site of
given the lack of consistent benefit found to date. inflammation. Because radiation-induced-lung injury
However, at least one group maintains that amifostine is characterized by inflammatory tissue reactions,
may produce clinically meaningful improvement in PGE2 and other prostaglandins as well as pro-
patient-reported symptoms (Movsas et al. 2005, 2009; inflammatory cytokines are produced in injured tissue
Dest 2006; Sarna et al. 2008). In one such analysis, in abundance. Because different prostanoids have
quality of life among 138 participants in RTOG 98-01 complementary or antagonistic activities, the final
was evaluated before and again 6 weeks after treat- biological effect on tissues depends on the balance of
ment for stages II–III NSCLC. Patients who had similar and opposing actions of the prostanoids
received amifostine reported greater pain reduction involved.
after treatment, less difficulty swallowing during Production of PGE2 and other pro-inflammatory
treatment, and less weight loss than patients who had prostanoids can be suppressed by non-steroidal anti-
not received amifostine. Interestingly, physician-rated inflammatory drugs (NSAIDs), which inhibit both
assessments of dysphagia were no different between isoforms of COX enzyme, or by selective COX-2
the two groups. The authors of this report concluded inhibitors. Because selective COX-2 inhibitors do not
that patient evaluations of difficulty in swallowing inhibit prostanoid production in normal tissues, they
and pain suggest benefits from amifostine that differ are less toxic than commonly used NSAIDs. Inter-
from clinician-rated assessments. estingly, both prostanoids and their inhibitors have
been reported to exert radioprotective actions on
normal tissues. Exogenous administration of PGE2,
3 Prostanoids, COX-2 and COX-2 other prostaglandins, or prostaglandin analogs before
Inhibitors irradiation of mice was shown to protect a variety of
tissues including hematopoietic tissue, jejunal
In response to physiological signals, stress, or injury mucosa, dermis, and testis (reviewed in Hanson 1998;
including radiation injury, cells produce prostanoids Milas and Hanson 1995). Prostaglandins vary widely
(prostaglandins and thromboxanes), a family of in their radioprotective ability; however, the prosta-
diverse, highly biologically active lipids derived from glandin analog misoprostol is amongst the most
the enzymatic metabolism of arachidonic acid by effective. Paradoxically, using NSAIDs to inhibit
cyclo-oxygenase (COX)-1 or COX-2. COX-1 is prostaglandins has also been shown to protect many
ubiquitous and responsible for prostanoid production tissues, including the lung, against radiation injury
in normal tissues, where prostanoids exert numerous (Milas and Hanson 1995; Michalowski 1994; Lee and
homeostatic physiological functions. In contrast, Stupans 2002). For example, Milas et al. (1992)
COX-2 is an inducible enzyme involved in prosta- reported that the NSAID indomethacin can protect
glandin production in pathologic states, particularly in mouse lung from radiation damage, but the protection
inflammatory processes and cancer. COX-2 is was limited to the early pneumonitis phase of injury.
induced by various factors including inflammatory Preliminary investigations of the selective COX-2
cytokines (such as tumor necrosis factor [TNF]-a, inhibitor SC-236 did not demonstrate significant
interleukin [IL]-1b, and platelet activity factors), protection from radiation-induced pneumonitis when
oncogenes, growth factors, and hypoxia. Prostanoids the drug was administered a few days before and after
participate in the pathogenesis of various pathologic lung irradiation. Subsequent experiments using a
states including inflammation; prostaglandin E2 different COX-2 inhibitor, celecoxib, provided sug-
(PGE2), a potent vasodilator and an immunosuppres- gestive evidence that giving the inhibitor during the
sive substance, is the major prostaglandin involved. development phase of acute pneumonitis may reduce
PGE2 is produced in abundance by pro-inflammatory either the latency or severity of lung injury. It should
232 R. Komaki et al.

be emphasized that even in the absence of lung (Mutter et al. 2009). This study was stopped early
radioprotection by COX-2 inhibitors, their adminis- because it did not meet the predetermined goal of an
tration can provide therapeutic benefit because of 80% response rate; findings from the evaluable
their potent enhancement of tumor radioresponse. The patients suggested that adding celecoxib to concurrent
ability of COX-2 inhibitors to selectively enhance chemoradiation did not improve survival. However,
tumor radioresponse has been reviewed in detail the findings did indicate that urinary levels of PGE-M,
elsewhere (Milas 2001, 2003b; Choy and Milas the major urinary metabolite of PGE2, may be useful
2003). as a marker for predicting response to celecoxib.
Another phase II study at the same institution com-
bining celecoxib with docetaxel chemotherapy for
3.1 Clinical Trials of COX-2 Inhibitors recurrent NSCLC (Csiki et al. 2005) produced an
for Lung Cancer overall response rate of 11% and a median survival
time of 6 months, similar to those observed after
Clinical and experimental findings have demonstrated docetaxel alone. The authors concluded that com-
conclusively that COX-2 overexpression correlates bining celecoxib with docetaxel at the doses and
with reduced survival among patients with NSCLC schedules used did not improve survival in unselected
(Khuri et al. 2001), that radiation treatment is asso- patients with recurrent previously treated NSCLC.
ciated with an increase in COX-2 expression in This disappointing finding was unexpected given
tumors (Steinauer et al. 2000), and that inhibiting ample preclinical evidence that celecoxib was highly
COX-2 activity in tumors is associated with an effective at enhancing tumor response to docetaxel
improved response to radiation (Choy and Milas (Nakata et al. 2004). A related clinical study, also
2003). Experimental evidence further indicates that underway at Vanderbilt, assessed celecoxib with
selective COX-2 inhibitors can enhance tumor radiation and taxane therapy for recurrent NSCLC;
response to radiation without substantially enhancing this combination produced two partial responses and
the radiosensitivity of normal tissues (Kishi et al. three disease stabilizations among 13 evaluable
2000). These findings have prompted several clinical patients, none of whom had responded to previous
trials combining selective COX-2 inhibitors with therapy. A fourth study at Vanderbilt is evaluating the
radiation for cancer treatment. These trials, results of benefit of radiation (62.5 Gy) combined with daily
which were mostly unpublished when this chapter celecoxib for inoperable stages I/II NSCLC. A phase I
was updated (Choy and Milas 2003; Baumann et al. study at MD Anderson Cancer Center (Liao et al.
2004; Komaki et al. 2004b), are discussed briefly 2005) evaluated the tolerability of celecoxib at doses
below. ranging from 200 to 800 mg/day given in combina-
The RTOG is conducting two trials using the tion with radiation for poor-prognosis NSCLC.
selective COX-2 inhibitor celecoxib with radiation, Among 47 enrolled patients, the main toxicities were
one a phase II study of postoperative celecoxib found to be grade 1 or 2 nausea and esophagitis, and
(400 mg twice daily) with radiation (50.4 Gy) for these were independent of the celecoxib dose or
patients with completely resected stages I/II NSCLC radiation schedule. Two patients, one given celecoxib
and the other a phase I/II trial of the same treatment 200 mg/day and the other at 400 mg/day, developed
for patients with stage IIB/III NSCLC treated with severe (grade 3) pneumonitis. Of 37 patients evalu-
fractionated radiation (66 Gy) and twice-daily cele- able for tumor response, 14 had complete response,
coxib for up to 2 years. Objectives in both studies 13 had partial responses, and 10 had stable or pro-
include assessing the tolerability of celecoxib at this gressive disease. Actuarial local progression-free
dose, evaluating the role of biomarkers as predictors survival rates of 66% at 1 year and 42% at 2 years
of celecoxib activity, and seeing if celecoxib were considered promising and worthy of further
improves the response and survival rates. assessment in a phase II/III trial.
Another phase II trial conducted at Vanderbilt Corticosteroids are highly potent anti-inflamma-
Cancer Center involved the addition of celecoxib to tory drugs used for symptomatic treatment of radia-
standard chemoradiation with paclitaxel and carbo- tion-induced pneumonitis. They inhibit production of
platin for previously untreated stage III NSCLC all prostanoids because, in addition to their ability to
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 233

inhibit COX enzymes, they prevent release of The radioprotective effect of b-FGF was attributed to
arachidonic acid from membrane phospholipids by its ability to increase cellular repair. A subsequent
stimulating the generation and secretion of lipocor- study by the same group (Fuks et al. 1994) showed
tins. (Ward et al. 1992a) showed that giving steroids that mice could be protected from lethal doses of
to rats at the time of radiation delivery protected rats whole lung irradiation if given intravenous b-FGF
from lung interstitial edema, delayed or suppressed immediately before or within 2 h after irradiation.
radiation-induced alveolitis, but did not affect devel- The effect was attributed to the protection of endo-
opment of pulmonary fibrosis. thelial cells against radiation-induced apoptosis. His-
tologic analysis of irradiated lung tissue, but not of
lungs exposed to both b-FGF and radiation, showed
4 Growth Factors and Cytokines apoptotic changes in the endothelial cell lining of the
pulmonary microvasculature within 6 to 8 h after
Growth factors and cytokines have critical roles in the radiation exposure. Also, histologic features of radi-
pathogenesis of radiation injury, including that to the ation-induced pneumonitis were absent in mice trea-
lung. Radiation alters the magnitude and dynamic ted with b-FGF. These results were not confirmed in a
activity of factors already present in affected tissues. subsequent study by Tee and Travis (1995) that
Response to radiation occurs within minutes or hours assessed the radioprotective action of b-FGF in two
after irradiation and can persist for days and months, strains of mice having different susceptibilities to
influencing the pathogenesis of both early and late radiation-induced lung injury. The reasons for the
radiation damage. Growth factors and cytokines discrepancy are unclear, but some differences in
principally act on cell and tissue proliferation, as well experimental conditions such as radiation dose, field
as cell loss. Hence, growth factors affect all the major size of radiation, and mouse strain could have
determinants of cell and tissue radioresponse: total accounted for this disparity.
number of clonogenic cells, cell cycle redistribution,
cell repopulation, cellular repair mechanisms, and
tissue microenvironment such as tumor hypoxia and 4.2 Keratinocyte Growth Factor
acidity. Many growth factors may be affected upon
tissue irradiation, those that have cytotoxic actions Keratinocyte growth factor (KGF) is another cell
and those that have cytoprotective ability, so that the growth factor that has been investigated for its ability
extent of tissue damage depends on the interaction to protect against radiation-induced lung damage.
of cytokines with similar or opposing activities. Although a member of the FGF family (FGF-7),
Involvement of growth factors and cytokines in the KGF’s cell growth stimulatory activity is confined to
pathogenesis of lung radiation damage is discussed in epithelial cells (Rubin et al. 1989; Miki et al. 1992).
more detail elsewhere in this volume. KGF was shown to be a good stimulator of the pro-
liferation of type II pneumocytes in vitro and in vivo
(Panos et al. 1993; Ulich et al. 1994), a type of cells
4.1 Basic Fibroblast Growth Factor considered to play an important role in repair of
injured lung tissue. Yi et al. (1996) showed that
Because some growth factors and cytokines may have intratracheal administration of KGF to rats 3 or
protective effects, attempts have been made to protect 2 days before the rats were exposed to 18 Gy
tissues that are at risk from lung cancer radiotherapy. of bilateral thoracic irradiation reduced the severity of
Basic fibroblast growth factor (b-FGF) was found radiation-induced pneumonitis and fibrosis observed
to protect endothelial cells from radiation both in histologically. However, no significant improvement
vitro (Haimovitz-Friedman et al. 1991) and in vivo was noted in rat survival. In contrast, KGF was highly
(Haimovitz-Friedman et al. 1991; Fuks et al. 1994). effective both in preventing development of bleomy-
To confer radiation resistance in vitro, b-FGF had to cin-induced fibrosis and in improving the survival of
be present at the time of radiation exposure or within treated animals. Significant protection was also ren-
several hours afterward. This protective effect was dered against the damage inflicted by the combination
abolished by treatment with anti-b-FGF antibodies. of bleomycin plus radiation treatment. A more recent
234 R. Komaki et al.

study (Terry et al. 2004) showed that a single intra- That study, which involved 93 patients (87 evaluable)
tracheal administration of recombinant human KGF with unresectable NSCLC and good performance status
to normal mice increased the proliferation of alveolar receiving 63 Gy in combination with weekly carbo-
epithelial cells 3 to 7 days later. This treatment platin and paclitaxel chemotherapy and 250 mg/m2 of
afforded significant protection against lethality from cetuximab, showed promising activity and median and
radiation-induced pneumonitis when the mice were overall survival times (22 and 24 months), longer than
irradiated at day 7 after administration of the growth any that have been previously reported by the RTOG.
factor. The efficacy of radiation may be further improved
by the concomitant administration of EGFR tyrosine
kinase inhibitors such as gefitinib and erlotinib with
4.3 Interleukin-11 radiation (Kim and Choy 2004). Combinations of
antiangiogenic agents such as bevacizumab with radi-
Regarding the radioprotective abilities of cytokines, it ation and chemotherapy have also produced encour-
is worth mentioning that subcutaneous administration aging results in glioma (Lai et al. 2011) and rectal
of recombinant interleukin-11 (rIL-11) rendered cancer (Willett et al. 2010). Combinations of agents
significant protection to mice from fatal thoracic that target both angiogenesis and EGFR signaling
irradiation (Redlich et al. 1996). The observed pathways, such as anti-VEGF antibodies (e.g., bev-
radioprotection was attributed to the rIL-11-induced acizumab) plus EGFR tyrosine kinase inhibitors, are
inhibition of radiation-induced expression of TNF being tested in clinical trials (Raben et al. 2004; Kerbel
mRNA as well as TNF production by macrophages. 2004). Currently, little evidence is available regarding
the synergy of molecular targeted drugs in combination
with radiation or chemoradiation for lung cancer, nor
4.4 Epidermal Growth Factor Receptor can definitive conclusions be drawn as to whether
adding a third agent (even a molecular targeted drug)
The epidermal growth factor receptor (EGFR) pathway improves the results compared with a standard two-
is associated with resistance to both cytotoxic chemo- drug regimen together with radiation (Eberhardt et al.
therapy and radiation therapy in cancer cell lines and is 2006). Local-regional control might be improved,
a validated therapeutic target in NSCLC (Benhar et al. however, by further dose intensification of radiation in
2002; Sumitomo et al. 2004; Liang et al. 2003; Goel this context (Bradley et al. 2005; Socinski et al. 2004).
et al. 2007; Shepherd et al. 2005). Cetuximab is an anti-
EGFR immunoglobulin G1 monoclonal antibody that
targets the extracellular domain of the EGFR, binding 5 Pentoxifylline
to it with a higher affinity than with the natural ligand
(Marshall 2006). Preclinical data indicate that cetux- Pentoxifylline (Trental), a methylxanthine derivative,
imab can amplify the response to chemotherapy and has is hemorrheologic agent that can reduce or ameliorate
radio sensitizing properties (Bruns et al. 2000; Baselga late radiation sequelae. In humans, pentoxifylline is
et al. 1993, 2000; Ciardiello et al. 1999; Raben et al. used to treat persistent soft tissue ulcerations and
2005; Saleh et al. 1999; Bianco et al. 2000; Milas necrosis. It has a variety of physiological activities
et al. 2000). Combinations of cetuximab with various including inhibition of platelet aggregation, regula-
types of chemotherapy for metastatic NSCLC have tion of tissue-damaging cytokines such as TNFa, and
shown that cetuximab is effective and tolerable with a enhancement of blood flow in injured microvascula-
manageable toxicity profile (Robert et al. 2005; Rosell ture. The drug may increase radioresponse of solid
et al. 2008; Lynch et al. 2010; Thienelt et al. 2005; tumors by increasing tumor oxygenation (Lee et al.
Pirker et al. 2009). These findings, in combination with 2000), and as such was tested in a clinical phase III
promising results from the concomitant use of cetux- trial in combination with radiotherapy for NSCLC
imab with radiation for head and neck cancer (Bonner (Kwon et al. 2000). That study showed that pentox-
et al. 2006) led to the initiation of a randomized ifylline was modestly effective, increasing the median
trial of cetuximab with chemoradiation for locally time to relapse by 2 months and the median survival
advanced stage III NSCLC (Blumenschein et al. 2011). time from 7 to 18 months. Although pentoxifylline
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 235

can modestly improve tumor radioresponse, it has against radiation-induced injury of several tissues
been used most often to reduce radiation injury to including the lung. In addition to inhibition of ACE,
normal tissues. Preclinical studies in experimental captopril is a free radical scavenger (Chopra et al.
animals have generally shown pentoxifylline to be 1989) and exhibits superoxide dismutase (SOD)-like
radioprotective, but the degree of protection varies activity (Roberts and Robinson 1995). Captopril
considerably. As an illustration, Lefaix et al. (1999) reduces radiation-induced pulmonary endothelial
reported striking regression of subcutaneous fibrosis dysfunction (Ward et al. 1988) and pulmonary fibrosis
induced by radiation to the skin surface of pigs using (Ward et al. 1990a, 1992c) and may ameliorate or
a combination of pentoxifylline and alpha-tocopherol delay radiation-induced pulmonary arterial hypoper-
(vitamin E). On the other hand, pentoxifylline seems fusion in rats (Graham et al. 1988; Ward et al. 1993).
to have little or no effect on acute skin or lung injuries Moreover, the use of ACE inhibitors as prophylaxis in
(Dion et al. 1989; Koh et al. 1995; Rube et al. 2002; rats receiving whole lung radiation was found to
Ward et al. 1992b). With respect to lung injury, reduce the radiation-induced activation of ACE and
pentoxifylline inhibited the radiation-induced plasminogen activator and reduce the production of
increase in TNFa mRNA during the acute phase of prostaglandins and thromboxane (Ward et al. 1988).
radiation injury, pneumonitis, but the impact of this Adding captopril to the feed after irradiation led to
biochemical change on lung injury was unclear (Rube reduced early lung reaction in rats receiving frac-
et al. 2002). In another study (Ward et al. 1992b), tionated hemithoracic irradiation (Ward et al. 1993).
pentoxifylline was found to further increase the In addition to pulmonary protection, ACE inhibition
radiation-induced production of prostanoids (PGI2 also protects against radiation injury of other tissues
and TXA2) while decreasing endothelial dysfunction including kidney (Moulder et al. 1993), skin (Ward
accompanied by increases in lung wet weight, protein et al. 1990b), jejunum (Yoon et al. 1994), and heart
and hydroxyproline content in the irradiated lung. (Yarom et al. 1993). With respect to the heart, Yarom
A randomized clinical trial, however, using prophy- et al. (1993) showed that captopril ameliorated the
lactic pentoxifylline showed a significant reduction in decrease in capillary function, increase in mast cells,
both early and late radiation-induced lung toxicities in fibrosis, number of atrial granules, and changes in
patients with breast or lung cancer (Ozturk et al. nerve terminals, but it failed to prevent the progres-
2004). A subsequent study of pentoxifylline with sive functional deterioration of the heart after
alpha-tocopherol given daily during and after radia- irradiation. Mechanisms of captopril-mediated radio-
tion therapy for lung cancer reduced the incidence of protection are not fully understood, but they are at
radiation-induced lung toxicity in both the subacute least partially related to its antihypertensive activity
and acute phases (Misirlioglu et al. 2007). The and its thiol-like function. Another study of captopril,
investigators suggested that this combination might enalopril (another ACE inhibitor), and an angiotensin
be especially useful for patients with lung cancer who II receptor blocker in rats receiving total-body irra-
receive concurrent chemoradiation therapy or have diation and cyclophosphamide as a model of bone
poor respiratory function. Other authors, however, marrow transplantation has implicated the inflamma-
have urged caution in interpreting the results of tory system as well (Molteni et al. 2007).
clinical trials of pentoxifylline, with or without vita- These promising preclinical observations on
min E, as many of those trials were nonrandomized or radioprotection by ACE inhibitors have led to several
had other design flaws (Nieder et al. 2005). clinical studies being undertaken. Wang et al. (2000)
reported a retrospective clinical study of ACE inhib-
itors given for the management of hypertension in
6 Angiotensin-Converting Enzyme patients with lung cancer treated with definitive
Inhibitors radiotherapy. The study showed that ACE inhibitors
given at a dose within the range used to treat hyper-
Angiotensin-converting enzyme (ACE) converts tension did not reduce the incidence or delay the onset
angiotensin I to angiotensin II, which is a potent of symptomatic radiation pneumonitis. A phase II
vasoconstrictor and hypertensive factor. Captopril is randomized clinical trial of captopril with radiation
an inhibitor of ACE that has been shown to protect and chemotherapy for lung cancer, RTOG 0123, was
236 R. Komaki et al.

closed early because of problems with accrual; the agents. In a preclinical study (Kim et al. 2004),
goal of that study was to see whether giving captopril treating mice bearing H460 lung carcinoma xeno-
after radiotherapy could reduce the incidence or grafts with docetaxel and flavopiridol significantly
severity of treatment-related pulmonary damage. enhanced the effects of radiation, with the effects
being greatest when docetaxel was given first, fol-
lowed by radiation, followed by the flavopiridol.
7 Flavopiridol As for clinical trials, most that have been completed
to date have tested flavopiridol as single-agent
Flavopiridol (alvocidib) is a synthetic cyclin-dependent therapy or in combination with chemotherapeutic
kinase inhibitor that has been shown in preclinical agents rather than with radiation (McInnes 2008).
studies to have both cytostatic and radiosensitizing Its effectiveness as a single-agent has been largely
effects in a variety of types of tumor cells, including disappointing in phase II trials of multiple myeloma
esophageal cancer (Sato et al. 2004; Raju et al. 2006), (Dispenzieri et al. 2006), melanoma (Burdette-Radoux
glioma (Newcomb et al. 2006), prostate cancer (Camp- et al. 2004), sarcoma (Morris et al. 2006), and endo-
hausen et al. 2004), and lung cancer (Kim et al. 2004). metrial adenocarcinoma (Grendys et al. 2005). More
Mason and colleagues at MD Anderson showed that encouraging results have been obtained from using
flavopiridol alone led to tumor growth delays in mice flavopiridol in combination with other drugs for
implanted with MCa-29 breast cancer cells, OCa-1 hematologic malignancies, such as cytarabine and
ovarian cancer cells, or Ly-TH lymphoma cells, but mitoxantrone for acute myelogenous leukemia (Karp
flavopiridol in combination with single-dose or frac- et al. 2007), fludarabine and rituximab for mantle-cell
tionated radiation led to significant radioenhancement, lymphoma, indolent B-cell non-Hodgkin’s lympho-
producing high rates of local tumor control or cure and, mas, and p53-mutated chronic lymphocytic leukemia
interestingly, antimetastatic activity as well (Mason (Lin et al. 2010), and cytosine arabinoside and
et al. 2004). These investigators concluded that thera- mitoxantrone for leukemias (Karp et al. 2007, 2010,
peutic gain was achieved when flavopiridol was initiated 2011). Combinations used to treat various solid tumors
either before or after the start of irradiation and that this have included flavopiridol with leucovorin, fluoroura-
compound showed promising clinical potential admin- cil, and oxaliplatin (FOLFOX) (Rathkopf et al. 2009);
istered alone or in combination with other cytotoxic with gemcitabine and irinotecan (Fekrazad et al. 2010);
agents. Raju et al. (2003, 2006), also at MD Anderson, with carboplatin or cisplatin (Bible et al. 2005); with
found that flavopiridol treatment significantly enhanced leucovorin, fluorouracil, and irinotecan (FOLFIRI)
the radiosensitivity of SEG-1 human esophageal ade- (Dickson et al. 2010); with carboplatin and paclitaxel
nocarcinoma cells (2006) and ovarian cancer cells (George et al. 2008); and with docetaxel (Carvajal et al.
(2003) in vitro and sharply enhanced the radioresponse 2009; Fornier et al. 2007; El-Rayes et al. 2006). The
of SEG-1 tumor xenografts (2006). Their findings fur- toxicity of flavopiridol is significant, consisting mostly
ther suggest that the mechanisms by which these effects of diarrhea and myelosuppression. Clearly, much work
take place include cell cycle redistribution, apoptosis, remains to be done to identify the most effective and
DNA repair, and transcriptional inhibition. Sato et al. least toxic regimens combining flavopiridol, radiation,
(2004) found that flavopiridol had a similar radiosensi- and other drugs for clinical applications in lung cancer.
tizing effect on three other esophageal squamous cell
carcinoma cell lines and also had dose-dependent effects
on cell cycle distribution and expression of cyclin D1, 8 Poly(ADP-Ribose) Polymerase
Rb, and Bcl-2 protein. Hara et al. (2008) also found Inhibitors
evidence of Bcl-2 involvement in their study of cervical
cancer cells; McAleer et al. (2006), studying a zebrafish As noted previously in this chapter, the responsive-
embryo model, found that the radiosensitizing effect of ness of tumor-cells to chemotherapy and radiation is
flavopiridol was attributable to inhibition of cyclin D1. determined by a combination of genetic factors, par-
In addition to its radiosensitizing properties, ticularly those that modify cell cycle arrest, repair of
flavopiridol has shown promise with regard to DNA damage, or cell death, and microenvironmental
potentiating the antitumor effects of chemotherapeutic factors such as hypoxia. Experimental evidence from
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 237

preclinical studies suggests that inhibitors to poly upregulated simultaneously by combining another
(ADP-ribose) polymerase (PARP), a nuclear enzyme Bcl-2 inhibitor, ABT-737, with rapamycin (Kim et al.
that recognizes and binds to DNA breaks to facilitate 2009). The combination of the two agents markedly
DNA strand break repair, may potentiate the effects of enhanced the sensitivity of H460 cells to radiation in
radiotherapy and chemotherapy. The radiosensitiza- clonogenic assays. Moreover, the combination of
tion effect of PARP inhibitors has been demonstrated ABT-737 and rapamycin and radiation led to drastic
in vitro in a broad variety of cancer cell cultures; tumor growth delays in a mouse xenograft model. The
moreover, this effect is present under both hypoxic combined treatment was confirmed to increase both
and euoxic conditions (Liu et al. 2008). PARP apoptosis and autophagy; it also suppressed tumor-
inhibitors have also shown radiosensitization cell proliferation and vascular density to a greater
in vivo in mouse xenograft models of cervical cancer extent than radiation alone. These investigators con-
(Kelland and Tonkin 1989), colon and rectal cancer cluded that concurrent induction of apoptosis and
(Donawho et al. 2007; Calabresi et al. 2004), head autophagy enhanced the effectiveness of radiation
and neck squamous cell carcinoma (Khan et al. 2010), therapy both in vitro and in lung cancer xenograft
glioblastoma (Russo et al. 2009), and lung cancer models, and suggested that the clinical potential of
(Albert et al. 2007). Although some clinical evidence this strategy should be explored for patients with lung
is emerging that supports the use of PARP inhibitors cancer.
with chemotherapy (Powell et al. 2010), such com-
binations have been associated with severe, dose-
limiting myelosuppression. No data have yet been 10 Efaproxaril
published from clinical trials of PARP inhibitors and
radiation therapy. Such trials should be designed with Efaproxaril (also known as RSR13) is a small syn-
special care given the substantial toxicity of these thetic molecule that noncovalently binds to hemo-
agents in combination with chemotherapy. globin, decreasing its oxygen-binding affinity and
shifting the oxygen dissociation curve to the left
(Viani et al. 2009). The effects of efaproxaril are
9 Bcl-2 Inhibitors based on increased oxygen levels in tumors, and thus
it can circumvent restrictions imposed by the blood–
Another potential molecular target for radiosensiti- brain barrier (Shaw et al. 2003; Kavanagh et al. 2001;
zation in lung cancer that has been the subject of Kunert et al. 1996; Suh 2004). Shaw et al. (2003)
attention is Bcl-2, a key regulator of apoptosis conducted a phase II, open-label, multicenter study of
thought to be responsible in part for the development efaproxaril plus whole-brain irradiation for 57
of radioresistance in tumor-cells. Moretti et al. (2010) patients with brain metastases and compared the
at Vanderbilt recently reported a preclinical study of results with those from the RTOG’s recursive parti-
the pan-Bcl-2 inhibitor AT-101 as a radiosensitizing tioning analysis (RPA) database on brain metastases.
agent in two lung cancer cell lines: A549, which is The mean survival time for patients treated with
radioresistant, and HCC2429, which is radiorespon- efaproxaril was slightly but significantly longer than
sive. AT-101 not only enhanced apoptosis in both cell that for the historical control group from the database
lines when used alone but also proved to be a potent (6.4 vs. 4.1 months, P \ 0.0174). In a subsequent
radiosensitizer for both radioresistant and radiore- phase III trial, Suh et al. (2006) also found a reduction
sponsive cell types. The authors concluded that Bcl-2 in risk of death and a modest improvement in survival
family members may serve as effective targets in lung associated with the use of efaproxaril in patients with
cancer and that clinical trials are warranted to assess brain metastases from breast or lung cancer, but the
the potential of AT-101 to enhance the therapeutic magnitude of the effect seemed more pronounced for
ratio of radiation therapy for lung cancer. the group with breast cancer. The addition of efa-
The same group also explored another strategy for proxaril to whole-brain irradiation for patients with
enhancing the radiosensitivity of H460 lung cancer brain metastases from breast cancer was also associ-
cells in which apoptosis and autophagy (degradation ated with improvements in quality of life and quality-
of cellular components via lysosomes) were adjusted survival (Scott et al. 2007).
238 R. Komaki et al.

organizing alveolitis (Epperly et al. 1999). Intratra-


11 Radioprotective Gene Therapy/ cheal MnSOD-PL gene therapy also reduced radiation-
Antioxidant Therapy: Superoxide induced inflammatory cytokines without rendering
Dismutase protection to orthotopic Lewis lung cancer models
(Guo et al. 2003). Preclinical animal studies suggested
The manganese superoxide dismutase (MnSOD) that radioprotective gene therapy reduces radiation-
located within the mitochondria is one of nature’s induced toxicity and may facilitate dose-escalation
most efficient catalysts. The enzyme protects redox protocols to improve the therapeutic ratio of lung
machinery within the mitochondria from the super- cancer radiotherapy. However, the efficacy and speci-
oxide radicals produced during normal respiration. In ficity of this approach need further investigation.
many pathological conditions, such as inflammation Application of MnSOD-PL gene therapy in the
caused by radiation-induced free radical damage, setting of fractionated chemoradiotherapy is being
superoxide is abundantly produced and may over- tested in clinical trials for prevention of esophagitis in
whelm the cell’s ability to efficiently remove it, patients with NSCLC. The gene therapy approach to
thereby leading to tissue injury. Via its antioxidant specifically deliver agents to targeted tissues is not
activity, MnSOD inactivates superoxide and hence limited to MnSOD but has high potential for delivery
has potential to protect against free radical induced of a wide array of agents including both cytotoxic and
injury. Early studies showed that systemic adminis- radioprotective agents.
tration of SOD can prevent radiation injury (Petkau
1987) and can even reduce preexisting radiation-
induced fibrosis (Delanian et al. 1994). When given 12 Concluding Remarks
before radiation, the activity of SOD has generally
been attributed to its radical scavenging effects, Normal tissue damage remains a major limiting factor
whereas when given after radiation the effects are in radiotherapy and chemoradiotherapy for cancer, as
most likely related to its anti-inflammatory and or improvements in tumor control and survival come at
immunostimulatory properties (Murray and McBride the cost of more common and more severe treatment-
1996; Grdina et al. 2009). Another SOD, recombinant related toxicity. For many years scientists have
CuZnSOD. was shown to protect the lung of hamsters explored various approaches to minimize damage to
from radiation-induced damage as evidenced by the tissues, including the use of chemical and biological
absence of severe histopathologic tissue changes 4 to radioprotective agents. As elaborated in this chapter,
16 weeks after irradiation and the prevention of ele- many of these agents have had significant radiopro-
vation of total protein content in brochoalveolar tective and chemoprotective effects in experimental
lavage (Breuer et al. 2000). animal models and some have been tested in clinical
More recently, a novel approach has been trials. Amifostine has been the most extensively
advanced for radioprotective gene therapy in which investigated, in both preclinical and clinical settings.
the antioxidant MnSOD is delivered to specific target Several clinical trials provided encouraging, although
organs such as lung and esophagus by gene transfer somewhat inconsistent, results with respect to reduc-
vectors such as plasmid/liposomes and adenovirus tions in the incidence and severity of esophagitis and
(Greenberger et al. 2003; Greenberger and Epperly pneumonitis. The agents discussed have complex
2007; Borrelli et al. 2009). Radiation protection by mechanisms of action and affect a variety of radia-
MnSOD transgene overexpression at the cellular level tion-induced tissue reactions both directly and indi-
is localized to the mitochondrial membrane. Intra- rectly. Radiation elicits the release of many
esophageal administration of MnSOD-PL before substances, such as growth factors, cytokines, and
irradiation induces transgene expression for 48–72 h, prostanoids, that can have both radioprotective and
with an associated decrease in radiation-induced radioenhancing properties. Because the final outcome
expression of inflammatory cytokine mRNA and of treatment depends on the balance between these
protein and esophagitis (Epperly et al. 2001, 2003). competing processes, the use of radioprotective
Intratracheal injection of adenovirus containing agents may act on only some of the many factors
MnSOD protected mice against radiation-induced involved. This is likely one reason for the
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 239

inconsistency in the literature on radioprotection. Bible KC, Lensing JL, Nelson SA et al (2005) Phase 1 trial of
Rapid achievements in recombinant technology and flavopiridol combined with cisplatin or carboplatin in
patients with advanced malignancies with the assessment
genetic engineering are opening possibilities to of pharmacokinetic and pharmacodynamic end points. Clin
upregulate or downregulate cellular expression of Cancer Res 11(16):5935–5941
diverse factors involved in tissue responses to radia- Blumenschein GR Jr, Paulus R, Curran WJ, Robert F, Fossella
tion and to select appropriate factors to achieve a F, Werner-Wasik M, Herbst R, Doescher PO, Choy H,
Komaki R (2011) A phase II study of cetuximab (C225) in
predetermined response. For example, redirecting the combination with chemoradiation in patients with stage
actions or optimizing the concentrations of a given IIIA/B non-small cell lung cancer: RTOG 0324 J Clin
response factor may become useful in increasing the Oncol 29(17):231–238
therapeutic ratio of radiotherapy by enhancing tumor Bonner HS, Shaw LM (2002) New dosing regimens for
amifostine: a pilot study to compare the relative bioavail-
radioresponse, or by reducing damage of normal tis- ability of oral and subcutaneous administration with
sues with radioprotectors. intravenous infusion. J Clin Pharmacol 42:166–174
Bonner JA, Harari PM, Giralt J et al (2006) Radiotherapy plus
cetuximab for squamous-cell carcinoma of the head and
References neck. N Engl J Med 354:567–578
Borrelli A, Schiattarella A, Mancini R et al (2009) A
recombinant MnSOD is radioprotective for normal cells
Albert JM, Cao C, Kim KW et al (2007) Inhibition of and radiosensitizing for tumor cells. Free Radic Biol Med
poly(ADP-ribose) polymerase enhances cell death and 46(1):110–116
improves tumor growth delay in irradiated lung cancer Bourhis J, Blanchard P, Maillard E et al (2011) On behalf of the
models. Clin Cancer Res 13(10):3033–3042 MAART Collaborative Group Effect of amifostine on
American Cancer Society (2011) Global Cancer Facts and survival among patients treated with radiotherapy: a meta-
Figures, 2nd edn. American Cancer Society, Atlanta analysis of individual patient data. J Clin Oncol (in press)
Antonadou D, Coliarakis N, Synodinou M et al (2001) Bradley J, Graham MV, Winter K et al (2005) Toxicity and
Randomized phase III trial of radiation treatment plus/ outcome results of RTOG 9311: a phase I–II dose escalation
minus amifostine in patients with advanced stage lung study using three-dimensional conformal radiation therapy
cancer. Int J Radiat Oncol Biol Phys 51:915–922 in patients with inoperable non-small cell lung cancinoma.
Antonadou D, Throuvalas N, Petridis A et al (2003) Effect of Int J Radiat Oncol Biol Phys 61:318–328
amifostine on toxicities associated with radiochemotherapy Breuer R, Tochner Z, Conner MW et al (2000) Superoxide
in patients with locally advanced non-small cell lung dismutase inhibits radiation-induced lung injury in ham-
cancer. Int J Radiat Oncol Biol Phys 57:402–408 sters. Lung 170:19–29
Bardet E, Martin L, Calais G et al (2002) Preliminary data of Brizel DM (2003) Does amifostine have a role in chemoradia-
the GORTEC 2000–02 phase III trial comparing intrave- tion treatment? Lancet Oncol 4:378–380
nous and subcutaneous administration of amifostine for Brizel DM (2007) Pharmacologic approaches to radiation
head and neck tumors treated by external radiotherapy. protection. J Clin Oncol 25(26):4084–4089
Semin Oncol 29:57–60 Brizel DM, Wasserman TH, Henke M et al (2000) Phase III
Baselga J, Norton L, Masui H et al (1993) Antitumor effects of randomized trial of amifostine as a radioprotector in head
doxorubicin in combination with anti-epidermal growth and neck cancer. J Clin Oncol 18:3339–3345
factor receptor monoclonal antibodies. J Natl Cancer Inst Bruns CJ, Harbison MT, Davis DW et al (2000) Epidermal
85:1327–1233 growth factor receptor blockade with C225 plus gemcita-
Baselga J, Pfister D, Cooper MR et al (2000) Phase I studies of bine results in regression of human pancreatic carcinoma
anti-epidermal growth factor receptor chimeric antibody growing orthotopically in nude mice by antiangiogenic
C225 alone and in combination with cisplatin. J Clin Oncol mechanisms. Clin Cancer Res 6:1936–1948
18:904–914 Burdette-Radoux S, Tozer RG et al (2004) Phase II trial of
Baumann M, Krause M, Zips D et al (2004) Molecular flavopiridol, a cyclin dependent kinase inhibitor, in
targeting in radiotherapy of lung cancer. Lung Cancer untreated metastatic malignant melanoma. Invest New
45(suppl 2):S187–S197 Drugs 22:315–322
Benhar M, Engelberg D, Levitzki A (2002) Cisplatin-induced Calabresi CR, Almassy R, Barton S et al (2004) Anticancer
activation of the EGF receptor. Oncogene 21:8723–8731 chemosensitization and radiosensitization by the novel
Bensadoun RJ, Schubert MM, Lalla RJ et al (2006) Amifostine poly(ADP-ribose) polymerase-1 inhibitor AG14361. J Natl
in the management of radiation-induced and chemo-induced Cancer Inst 96(1):56–67
mucositis. Support Care Cancer 14:566–572 Calabro-Jones PM, Gahey RC, Smoluk GD et al (1985)
Bianco C, Bianco R, Tortora G et al (2000) Antitumor activity Alkaline phosphatase promotes radioprotection and accu-
of combined treatment of human cancer cells with ionizing mulation of WR-1065 in V79–171 cells incubated in
radiation and anti-epidermal growth factor receptor mono- medium containing WR-2721. Int J Radiat Biol 47:23–27
clonal antibody C225 plus type I protein kinase A antisense Camphausen K, Brady KJ, Burgan WE et al (2004) Flavopir-
oligonucleotide. Clin Cancer Res 6:4343–4350 idol enhances human tumor cell radiosensitivity and
240 R. Komaki et al.

prolongs expression of gammaH2AX foci. Mol Cancer Ther Eberhardt W, Pöttgen C, Stuschke M (2006) Chemoradiation
3(4):409–416 paradigm for the treatment of lung cancer. Nat Clin Pract
Carvajal RD, Tse A, Shah MA et al (2009) A phase II study of Oncol 3(4):188–199
flavopiridol (Alvocidib) in combination with docetaxel in El-Rayes BF, Gadgeel S, Parchment R et al (2006) A phase I
refractory, metastatic pancreatic cancer. Pancreatology study of flavopiridol and docetaxel. Invest New Drugs
9(4):404–409 24(4):305–310
Chopra M, Scott N, McMurray J et al (1989) A free radical Epperly MW, Bray JA, Krager S et al (1999) Intratracheal
scavenger. Br J Clin Pharmacol 27:396–399 injectrion of adenovirus containing the human MnSOD
Choy H, Milas L (2003) Enhancing radiotherapy with transgene protects athymic nude mice from irradiation-
cyclooxygenase-2 enzyme inhibitors: a rational advance? J induced organizing alveolitis. Int J Radiat Oncol Biol Phys
Natl Cancer Inst 95:1440–1452 43:169–181
Ciardiello F, Bianco R, Damiano V et al (1999) Antitumor Epperly MW, Gretton JA, DeFilippi SJ et al (2001) Modulation
activity of sequential treatment with topotecan and anti- of radiation-induced cytokine elevation associated with
epidermal growth factor receptor monoclonal antibody esophagitis and esophageal stricture by manganese super-
C225. Clin Cancer Res 5:909–916 oxide dismutase-plasmid/liposome (SOD2-PL) gene ther-
Csiki I, Morrow JD, Sandler A et al (2005) Targeting apy. Radiat Res 155:2–14
cyclooxygenase-2 in recurrent non-small cell lung cancer: Epperly MW, Guo HL, Jefferson M et al (2003) Cell phenotype
a phase II trial of celecoxib and docetaxel. Clin Cancer Res specific kinetics of expression of intratracheally injected
11(18):6634–6640 manganese superoxide dismutase plasmid/liposomes
Curran WJ, Scott CB, Langer CJ et al (2003) Long-term benefit (MnSOD-PL) during lung radioprotective gene therapy.
is observed in a phase III comparison of sequential versus Gene Ther 2:163–171
concurrent chemo-radiation for patients with unresected Fekrazad HM, Verschraegen CF, Royce M et al (2010) A phase
stage III NSCLC: RTOG 9410. Proc Am Soc Clin Oncol I study of flavopiridol in combination with gemcitabine and
22:621 (abstr 2499) irinotecan in patients with metastatic cancer. Am J Clin
Delanian S, Baillet F, Huart J et al (1994) Successful treatment Oncol 33(4):393–397
of radiation-induced fibrosis using liposomal Cu/Zn super- Fornier MN, Rathkopf D, Shah M et al (2007) Phase I dose-
oxide dismutase: clinical trial. Radiother Oncol finding study of weekly docetaxel followed by flavopiridol
32:12–20 for patients with advanced solid tumors. Clin Cancer Res
Dest VM (2006) Radioprotectants: adding quality of life to 13(19):5841–5846
survivorship? Semin Oncol Nurs 22(4):249–256 Fuks Z, Persaud RS, Alfieri A et al (1994) Basic fibroblast
Dickson MA, Shah MA, Rathkopf D et al (2010) A phase I growth factor protects endothelial cells against radiation-
clinical trial of FOLFIRI in combination with the induced programmed cell death in vitro and in vivo. Cancer
pan-cyclin-dependent kinase (CDK) inhibitor flavopiridol. Res 54:2582–2590
Cancer Chemother Pharmacol 66(6):1113–1121 George S, Kasimis BS, Cogswell J et al (2008) Phase I study of
Dillman RO, Herndon J, Seagren SL et al (1996) Improved flavopiridol in combination with Paclitaxel and Carboplatin
survival in stage III non-small-cell lung cancer: seven-year in patients with non-small-cell lung cancer. Clin Lung
follow-up of Cancer and Leukemia Group B (CALGB) Cancer 9(3):160–165
8433 trial. J Natl Cancer Inst 88:1210–1215 Giannopoulou E, Papadimitriou E (2003) Amifostine has antian-
Dillman RO, Seagren SL, Propert KJ et al (1990) A randomized giogenic properties in vitro by changing the redox status of
trial of induction chemotherapy plus high-dose radiation human endothelial cells. Free Radic Res 37:1191–1199
versus radiation alone in stage III non-small-cell lung Goel S, Hidalgo M, Perez-Soler R (2007) EGFR inhibitor-
cancer. N Engl J Med 323:940–945 mediated apoptosis in solid tumors. J Exp Ther Oncol
Dion MW, Hussey DH, Osborne JW (1989) The effect of 6:305–320
pentoxifylline on early and late radiation injury following Gopal R, Starkschall G, Tucker SL et al (2003) Effects of
fractionated irradiation of C3H mice. Int J Radiat Oncol radiotherapy and chemotherapy on lung function in patients
Biol Phys 17:101–107 with non-small-cell lung cancer. Int J Radiat Oncol Biol
Dispenzieri A, Gertz MA, Lacy MQ et al (2006) Flavopiridol in Phys 56(1):114–120
patients with relapsed or refractory multiple myeloma: a Graham NN, Evans ML, Dahlen DD et al (1988) Drug
phase 2 trial with clinical and pharmacodynamic end-points. suppression of late radiation injury in the rat lung.
Haematologica 91:390–393 philadelphia: 36th annual meeting of the radiation research
Donawho CK, Luo Y, Penning TD et al (2007) ABT-888, an society, April 16–21
orally active poly(ADP-ribose) polymerase inhibitor that Grdina DJ, Kataoka Y, Murley JS et al (2002) Inhibition of
potentiates DNA-damaging agents in preclinical tumor spontaneous metastases formation by amifostine. Int J
models. Clin Cancer Res 13(9):2728–2737 Cancer 97:135–141
Durand RE (1983) Radioprotection by WR-2721 in vitro and Grdina DJ, Murley JS, Kataoka Y et al (2009) Amifostine
low oxygen tensions: implications for its mechanisms of induces antioxidant enzymatic activities in normal tissues
action. Br J Cancer 47:387–392 and a transplantable tumor that can affect radiation
Durand RE, Olive PL (1989) Radiosensitization and radio- response. Int J Radiat Oncol Biol Phys 73(3):886–896
protection by BSO and WR-2721: the role of oxygenation. Greenberger JS, Epperly MW, Gretton J et al (2003) Radio-
Br J Cancer 60:417–522 protective gene therapy. Curr Gene Ther 3:183–195
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 241

Greenberger JS, Epperly MW (2007) Antioxidant gene doses and correlation of pharmacokinetics with pharmaco-
therapeutic approaches to normal tissue radioprotection dynamics. Int J Radiat Oncol Biol Phys 49:1133–1139
and tumor radiosensitization. In Vivo 21:141–146 Kelland LR, Tonkin KS (1989) The effect of 3-aminobenza-
Grendys EC Jr, Blessing JA, Burger R et al (2005) A phase II mide in the radiation response of three human cervix
evaluation of flavopiridol as second-line chemotherapy of carcinoma xenografts. Radiother Oncol 15(4):363–369
endometrial carcinoma: a Gynecologic Oncology Group Kemp G, Rose P, Lurain J et al (1996) Amifostine preatreat-
Study. Gynecol Oncol 98:249–253 ment for protection against cyclophosphamide-induced and
Guo H, Epperly MW, Bernarding M et al (2003) Manganese cisplatin-induced toxicities: results of a randomized control
superoxide dismutase-plasmid/liposome (MnSOD-PL) trial in patients with advanced ovarian cancer. J Clin Oncol
intratracheal gene therapy reduction of irradiation induced 14:2101–2112
inflammatory cytokines does not protect orthotopic Lewis Kerbel RS (2004) Antiangiogenic drugs and current strategies
lung carcinomas. In Vivo 17:13–21 for the treatment of lung cancer. Semin Oncol 31(1
Haddad R, Wirth L, Costello R et al (2003) Phase II suppl):54–60
randomized study of concomitant chemoradiation using Khan K, Araki K, Wang D et al (2010) Head and neck cancer
weekly carboplatin/paclitaxel with or without daily subcu- radiosensitization by the novel poly(ADP-ribose) polymer-
taneous amifostine in patients with newly diagnosed locally ase inhibitor GPI-15427. Head Neck 32(3):381–391
advanced squamous cell carcinoma of the head and neck. Khuri FR, Wu H, Lee JJ et al (2001) Cyclooxygenase-2
Semin Oncol 30(6 suppl 18):84–88 overexpression is a marker of poor prognosis in stage I non-
Haimovitz-Friedman A, Vlodavsky I, Chaudhuri A et al (1991) small cell lung cancer. Clin Cancer Res 7:861–867
Autocrine effects of fibroblast growth factor in repair of Kim DW, Choy H (2004) Potential role for epidermal growth
radiation damage in endothelial cells. Cancer Res 51: factor receptor inhibitors in combined-modality therapy for
2552–2558 non-small-cell lung cancer. Int J Radiat Oncol Biol Phys
Han HS, Han JY, Yu SY et al (2008) Randomized phase 2 59(2 suppl):S11–S20
study of subcutaneous amifostine versus epoetin-alpha Kim JC, Saha D, Cao Q et al (2004) Enhancement of radiation
given 3 times weekly during concurrent chemotherapy and effects by combined docetaxel and flavopiridol treatment in
hyperfractionated radiotherapy for limited-disease small lung cancer cells. Radiother Oncol 71(2):213–221
cell lung cancer. Cancer 113:1623–1631 Kim KW, Moretti L, Mitchell LR et al (2009) Combined Bcl-2/
Hanson W (1998) Eicosanoid-induced radioprotection and mammalian target of rapamycin inhibition leads to
chemoprotection: laboratory studies and clinical applica- enhanced radiosensitization via induction of apoptosis and
tions. In: Bump E, Malaker K (eds) Radioprotectors: autophagy in non-small cell lung tumor xenograft model.
chemical, biological and clinical perspectives. CRC Press, Clin Cancer Res 15(19):6096–6105
Boca Raton, pp 197–221 Kishi K, Petersen S, Petersen C et al (2000) Preferential
Hara T, Omura-Minamisawa M, Kang Y et al (2008) Flavo- enhancement of tumor radioresponse by a cyclooxygenase-2
piridol potentiates the cytotoxic effects of radiation in inhibitor. Cancer Res 60:1326–1331
radioresistant tumor cells in which p53 is mutated or Bcl-2 Kligerman MM, Turrisi AT, Urtasan RC et al (1988) Final
is overexpressed. Int J Radiat Oncol Biol Phys 71(5): report on phase I trial of WR-2721 before protracted
1485–1495 fractionated radiation therapy. Int J Radiat Oncol Biol Phys
Hensley ML, Hagerty KL, Kewalramani T et al (2009) 14:1119–1122
American Society of Clinical Oncology 2008 clinical Koh WJ, Stelzer KJ, Peterson LM et al (1995) Effect of
practice guideline update: use of chemotherapy and radia- pentoxifylline on radiation-induced lung and skin toxicity in
tion therapy protectants. J Clin Oncol 27:127–145 rats. Int J Radiat Oncol Biol Phys 31:71–77
Hunter N, Milas L (1983) Protection by S-2- (3-Aminopropy- Komaki R, Lee JS, Milas L et al (2004a) Effects of amifostine
lamino)- ethylphosphorothioic acid against radiation- on acute toxicity from concurrent chemotherapy and
induced leg contractures in mice. Cancer Res 43:1630–1632 radiotherapy for inoperable non-small cell lung cancer:
Jemal A, Tiwari RC, Murray T et al (2004) Cancer statistics, report of a randomized comparative trial. Int J Radiat Oncol
2004. CA Cancer J Clin 54:8–29 Biol Phy 58:1369–1377
Karp JE, Blackford A, Smith BD et al (2010) Clinical activity Komaki R, Liao Z, Milas L (2004b) Improvement strategies for
of sequential flavopiridol, cytosine arabinoside, and molecular targeting: cyclooxygenase-2 inhibitors as radio-
mitoxantrone for adults with newly diagnosed, poor-risk sensitizers for non-small cell lung cancer. Semin Oncol 31(1
acute myelogenous leukemia. Leuk Res 34(7):877–882 suppl 1):47–53
Karp JE, Smith BD, Resar LS et al (2011) Phase I and Komaki R, Seiferheld W, Ettinger D et al (2002) Randomized
pharmacokinetic study of bolus-infusion flavopiridol fol- phase II chemotherapy and radiotherapy trial for patients
lowed by cytosine arabinoside and mitoxantrone for acute with locally advanced inoperable non-small-cell lung can-
leukemias. Blood 117(12):3302–3310 cer: long-term follow-up of RTOG 92–04. Int J Radiat
Karp JE, Smith BD, Levis MJ et al (2007) Sequential Oncol 53:548–557
flavopiridol, cytosine arabinoside, and mitoxantrone: a Koukourakis MI, Kyrias G, Kakolyris S et al (2000) Subcu-
phase II trial in adults with poor-risk acute myelogenous taneous administration of amifostine during fractionated
leukemia. Clin Cancer Res 13:4467–4473 radiotherapy: a randomized Phase II study. J Clin Oncol
Kavanagh BD, Khandelwal SR, Schmidt-Ullrich RK et al 18:2226–2233
(2001) A phase I study of RSR13, a radiation-enhancing Koukourakis MI, Simopoulos C, Minopoulos G et al (2003)
hemoglobin modifier: tolerance of repeated intravenous Amifostine before chemotherapy: improved tolerance
242 R. Komaki et al.

profile of the subcutaneous over the intravenous route. Clin Marzatico F, Porta C, Moroni M et al (2000) In vitro
Cancer Res 15:3288–3293 antioxidant properties of amifostine (WR-2721, EthyolTM).
Kouvaris JR, Kouloulias VE, Vlahos LJ (2007) Amifostine: the Cancer Chemother Pharmacol 45:172–176
first selective-target and broad-spectrum radioprotector. Mason KA, Hunter NR, Raju U et al (2004) Flavopiridol
Oncologist 12:738–747 increases therapeutic ratio of radiotherapy by preferentially
Kunert MP, Liard JF, Abraham DJ (1996) RSR-13, an allosteric enhancing tumor radioresponse. Int J Radiat Oncol Biol
effector of hemoglobin, increases systemic and iliac Phys 59(4):1181–1189
vascular resistance in rats. Am J Physiol 271:H602–H613 McAleer MF, Duffy KT, Davidson WR et al (2006) Antisense
Kwon HC, Kim SK, Chung WK et al (2000) Effect of inhibition of cyclin D1 expression is equivalent to flavo-
pentoxifylline on radiation response of non-small cell lung piridol for radiosensitization of zebrafish embryos. Int J
cancer: a phase III randomized multicenter trial. Radiother Radiat Oncol Biol Phys 66(2):546–551
Oncol 56:175–179 McInnes C (2008) Progress in the evaluation of CDK inhibitors
Lai A, Tran A, Nghiemphu PL et al (2011) Phase II study of as antitumor agents. Drug Discov Today 13:875–881
bevacizumab plus temozolomide during and after radiation Mell LK, Malik R, Komaki R et al (2007) Effect of amifostine
therapy for patients with newly diagnosed glioblastoma on response rates in locally advanced non-small-cell lung
multiforme. J Clin Oncol 29(2):142–148 cancer patients treated on randomized controlled trials: a
LeChevalier T, Arriagada R, Quoix E et al (1991) Radiotherapy meta-analysis. Int J Radiat Oncol Biol Phys 68:111–118
alone versus combined chemotherapy and radiotherapy in Michalowski AS (1994) On radiation damage to normal tissues
nonresectable non-small-cell lung cancer: first analysis of a and its treatment: II. Anti-inflammatory drugs. Acta Oncol
randomized trial in 353 patients. J Natl Cancer Inst 83:417–423 33:139–157
Lee I, Biaglow JE, Lee J et al (2000) Physiological mechanisms Miki T, Bottaro DP, Fleming TP et al (1992) Determination of
of radiation sensitization by pentoxifylline. Anticancer Res ligand-binding specificity by alternative splicing: two
20(6B):4605–4609 distinct growth factor receptors encoded by a single gene.
Lee TK, Stupans I (2002) Radioprotection: the non-steroidal Proc Natl Acad Sci USA 89:246–250
anti-inflammatory drugs (NSAIDs) and prostaglandins. Milas L (2001) Cyclooxygenase-2 (COX-2) enzyme inhibitors
J Pharm Pharmacol 54:1435–1445 as potential enhancers of tumor radioresponse. Semin
Lefaix JL, Delanian S, Vozenin MC et al (1999) Striking Radiat Oncol 11:290–299
regression of subcutaneous fibrosis induced by high doses of Milas L, Hanson WR (1995) Eicosanoids and radiation. Eur J
gamma rays using a combination of pentoxifylline and a- Cancer 31A:1580–1585
tocopherol: an experimental study. Int J Radiat Oncol Biol Milas L, Hunter N, Reid BO et al (1982) Protective effects of S-
Phys 43:839–847 2-(3-Aminopropylamino)-ethylphosphorothioic acid against
Leong SS, Tan EH, Fong KW et al (2003) Randomized double- radiation damage of normal tissues and a fibrosarcoma in
blind trial of combined modality treatment with or without mice. Cancer Res 42:1888–1897
amifostine in unresectable stage III non-small cell lung Milas L, Mason K, Hunter N et al (2000) In vivo enhancement
cancer. J Clin Oncol 21:1767–1774 of tumor radioresponse by C225 antiepidermal growth
Liang K, Ang KK, Milas L et al (2003) The epidermal growth factor receptor antibody. Clin Cancer Res 6:701–708
factor receptor mediates radioresistance. Int J Radiat Oncol Milas L, Mason KA, Liao Z et al (2003a) Chemoradiotherapy:
Biol Phys 57:246–254 emerging treatment improvement strategies. Head Neck
Liao Z, Komaki R, Milas L et al (2005) A phase I clinical trial 25:152–167
of thoracic radiotherapy and concurrent celecoxib for Milas L, Mason K, Liao Z et al (2003b) Role of cyclooxygenase-
patients with unfavorable performance status inoperable/ 2 (COX-2) and its inhibition in tumor biology and radio-
unresectable non-small cell lung cancer. Clin Cancer Res therapy. In: Nieder C, Milas L, Ang KK (eds) Biological
11:3342–3348 modification of radiation response: cytokines, growth factors
Lin TS, Blum KA, Fischer DB et al (2010) Flavopiridol, and other biological targets. Springer, Berlin, pp 241–258
fludarabine, and rituximab in mantle cell lymphoma and Milas L, Murray D, Brock WA et al (1988) Radioprotectors in
indolent B-cell lymphoproliferative disorders. J Clin Oncol tumor radiotherapy: factors and settings determining ther-
28(3):418–423 apeutic ratio. Pharmacol Ther 30:179–187
Liu SK, Coackley C, Krause M et al (2008) A novel poly(ADP- Milas L, Nishiguchi I, Hunter N et al (1992) Radiation
ribose) polymerase inhibitor, ABT-888, radiosensitizes protection against early and late effects of ionizing irradi-
malignant human cell lines under hypoxia. Radiother Oncol ation by the prostaglandin inhibitor indomethacin. Adv
88(2):258–268 Space Res 12:265–271
Lockhart SP (1990) Inhaled thiol and phosphothiol radio- Misirlioglu CH, Demirkasimoglu T, Kucukplakci B et al (2007)
protectors fail to protect the mouse lung. Radiother Oncol Pentoxifylline and alpha-tocopherol in prevention of radi-
19:187–191 ation-induced lung toxicity in patients with lung cancer.
Lynch T, Patel T, Dreisbach L et al (2010) Cetuximab and Med Oncol 24(3):308–311
first-line taxane/carboplatin chemotherapy in advanced non- Molteni A, Wolfe LF, Ward WF et al (2007) Effect of an
small-cell lung cancer: results of the randomized multicen- angiotensin II receptor blocker and two angiotensin con-
ter phase III trial BMS099. J Clin Oncol 28(6):911–917 verting enzyme inhibitors on transforming growth factor-
Marshall J (2006) Clinical implications of the mechanism of beta (TGF-beta) and alpha-actomyosin (alpha SMA),
epidermal growth factor receptor inhibitors. Cancer 107: important mediators of radiation-induced pneumopathy
1207–1218 and lung fibrosis. Curr Pharm Des 13(13):1307–1316
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 243

Moretti L, Li B, Kim KW et al (2010) AT-101, a pan-Bcl-2 preclinical results from a radiation oncology perspective.
inhibitor, leads to radiosensitization of non-small cell lung Int J Radiat Oncol Biol Phys 59(2 Suppl):S27–S38
cancer. J Thorac Oncol 5(5):680–687 Raben D, Helfrich B, Chan DC et al (2005) The effects of
Morris DG, Bramwell VH, Turcotte R et al (2006) A phase II cetuximab alone and in combination with radiation and/or
study of flavopiridol in patients with previously untreated chemotherapy in lung cancer. Clin Cancer Res 11:795–805
advanced soft tissue sarcoma. Sarcoma 2006:64374 Raju U, Ariga H, Koto M et al (2006) Improvement of
Moulder JE, Fish BL, Cohen EP (1993) Treatment of radiation esophageal adenocarcinoma cell and xenograft responses to
nephropathy with ACE inhibitors. Int J Radiat Oncol Biol radiation by targeting cyclin-dependent kinases. Radiother
Phys 27:93–99 Oncol 80(2):185–191
Movsas B, Moughan J, Sarna L et al (2009) Quality of life Raju U, Nakata E, Mason KA et al (2003) Flavopiridol, a
supersedes the classic prognosticators for long-term survival cyclin-dependent kinase inhibitor, enhances radiosensitivity
in locally advanced non-small-cell lung cancer: an analysis of ovarian carcinoma cells. Cancer Res 63(12):3263–3267
of RTOG 9801. J Clin Oncol 27(34):5816–5822 Rasey JS, Grunbaum Z, Krohn KA et al (1985) Biodistribution of
Movsas B, Scott C, Langer C et al (2005) Randomized trial of the radioprotective drug 35S-labeled 3-amino-2-hydroxypro-
amifostine in locally advanced non–small-cell lung cancer pyl phosphorothioate (WR77913). Radiat Res 102:130–137
patients receiving chemotherapy and hyperfractionated Rasey JS, Krohn KA, Menard TW et al (1986) Comparative
radiation: radiation therapy oncology group trial 98–01. biodistribution and radioprotection studies with three
J Clin Oncol 23:2145–2154 radioprotective drugs in mouse tumors. Intl J Radiat Oncol
Murray D, McBride WH (1996) Radioprotective agents. In: Biol Phys 12:1487–1490
Kirk-Othmer Encyclopedia of Chemical Technology, 4th Rathkopf D, Dickson MA, Feldman DR et al (2009) Phase I
edn. vol 20, pp. 963–1006 study of flavopiridol with oxaliplatin and fluorouracil/
Mutter R, Lu B, Carbone DP et al (2009) A phase II study of leucovorin in advanced solid tumors. Clin Cancer Res
celecoxib in combination with paclitaxel, carboplatin, and 15(23):7405–7411
radiotherapy for patients with inoperable stage IIIA/B non- Redlich CA, Gao X, Rockwell S et al (1996) IL-11 enhances
small cell lung cancer. Clin Cancer Res 15(6):2158–2165 survival and decreases TNF production after radiation-
Nakata E, Mason KA, Hunter N et al (2004) Potentiation of induced thoracic injury. J Immunol 157:1705–1710
tumor response to radiation or chemoradiation by selective Robert F, Blumenschein G, Herbst RS et al (2005) Phase I/IIa
cyclooxygenase-2 enzyme inhibitors. Int J Radiat Oncol study of cetuximab with gemcitabine plus carboplatin in
Biol Phys 58(2):369–375 patients with chemotherapy-naive advanced non-small-cell
Newcomb EW, Lymberis SC, Lukyanov Y et al (2006) lung cancer. J Clin Oncol 23:9089–9096
Radiation sensitivity of GL261 murine glioma model and Roberts NA, Robinson PA (1995) Copper chelates of anti-
enhanced radiation response by flavopiridol. Cell Cycle rheumatic and anti-inflammatory agents and their super-
5(1):93–99 oxide dismutase-like activity and stability. Br J Rheumatol
Nieder C, Zimmermann FB, Adam M et al (2005) The role of 24:128–136
pentoxifylline as a modifier of radiation therapy. Cancer Rosell R, Robinet G, Szczesna A et al (2008) Randomized
Treat Rev 31(6):448–455 phase II study of cetuximab plus cisplatin/vinorelbine
O’Rourke N, Roqué I, Figuls M et al (2010) Concurrent compared with cisplatin/vinorelbine alone as first-line
chemoradiotherapy in non-small cell lung cancer. Cochrane therapy in EGFR-expressing advanced non-small-cell lung
Database Syst Rev 16(6):CD002140 cancer. Ann Oncol 19:362–369
Ozturk B, Egehan I, Atavci S et al (2004) Pentoxifylline in Rube CE, Wilfert F, Uthe D et al (2002) Modulation of
prevention of radiation-induced lung toxicity in patients radiation-induced tumour necrosis factor alpha (TNF-alpha)
with breast and lung cancer: a double-blind randomized expression in the lung tissue by pentoxifylline. Radiother
trial. Int J Radiat Oncol Biol Phys 58:213–219 Oncol 64:177–187
Panos RJ, Rubin JS, Aaronson SA et al (1993) Keratinocyte Rubin JS, Osada H, Finch PW et al (1989) Purification and
growth factor and hepatocyte growth factor/scatter factor characterization of a newly identified growth factor specific
are heparin-binding growth factors for alveolar type II cells for epithelial cells. Proc Natl Acad Sci USA 86(3):802–806
in fibroblast-conditioned medium. J Clin Invest 92:969–977 Russo AL, Kwon HC, Burgan WE et al (2009) In vitro and in
Petkau A (1987) Role of superoxide dismutase in modification vivo radiosensitization of glioblastoma cells by the poly
of radiation injury. Br J Cancer 55(suppl VIII):87–95 (ADP-ribose) polymerase inhibitor E7016. Clin Cancer Res
Pirker R, Pereira JR, Szczesna A et al (2009) Cetuximab plus 15(2):607–612
chemotherapy in patients with advanced non-small-cell lung Saleh MN, Raisch KP, Stackhouse MA et al (1999) Combined
cancer (FLEX): an open-label randomised phase III trial. modality therapy of A431 human epidermoid cancer using
Lancet 373:1525–1531 anti-EGFr antibody C225 and radiation. Cancer Biother
Powell C, Mikropoulos C, Kaye SB et al (2010) Preclinical and Radiopharm 14:451–463
clinical evaluation of PARP inhibitors as tumour-specific Sarna L, Swann S, Langer C et al (2008) Clinically meaningful
radiosensitisers. Cancer Treat Rev 36(7):566–575 differences in patient-reported outcomes with amifostine in
Praetorius NP, Mandal TK (2008) Alternate delivery route for combination with chemoradiation for locally advanced non-
amifostine as a radio-/chemo-protecting agent. J Pharm small-cell lung cancer: an analysis of RTOG 9801. Int J
Pharmacol 60:809–815 Radiat Oncol Biol Phys 72:1378–1384
Raben D, Helfrich B, Bunn PA Jr (2004) Targeted therapies Sasse AD, Clark LG, Sasse EC et al (2006) Amifostine reduces
for non-small-cell lung cancer: biology, rationale, and side effects and improves complete response rate during
244 R. Komaki et al.

radiotherapy: results of a meta-analysis. Int J Radiat Oncol Thienelt CD, Bunn PA Jr, Hanna N et al (2005) Multicenter
Biol Phys 64:784–791 phase I/II study of cetuximab with paclitaxel and carbo-
Sato S, Kajiyama Y, Sugano M et al (2004) Flavopiridol as a platin in untreated patients with stage IV non-small-cell
radio-sensitizer for esophageal cancer cell lines. Dis lung cancer. J Clin Oncol 23:8786–8793
Esophagus 17(4):338–344 Travis E (1984) The oxygen dependence of protection by
Savoye C, Swenberg C, Hugot S (1997) Thiol WR-1065 and aminothiols: implications for normal tissues and solid
disulphide WR-33278, two metabolities of the drug ethyol tumors. Int J Radiat Oncol Biol Phys 10:1495–1501
(WR-2721), protect DNA against fast neutron-induced Travis EL, Thames HD Jr, Tucker SL et al (1985) Late
strand breakage. Int J Radiat Biol 71:193–202 functional and biochemical changes in mouse lung after
Schaake-Konig C, van den Bogaert W, Dalesio O et al (1992) irradiation: differential effects of WR-2721. Rad Res 103:
Effects of concomitant cisplatin and radiotherapy on 219–231
inoperable non-small-cell lung cancer. N Engl J Med 326: Turrisi AT, Kim K, Blum R et al (1999) Twice-daily compared
524–530 with once-daily thoracic radiotherapy in limited small-cell
Schuchter LM, Glick J (1993) The current status of WR-2721 lung cancer treated concurrently with cisplatin and etopo-
(amifostine): a chemotherapy and radiation therapy protec- side. N Engl J Med 340:265–271
tor. J Clin Oncol 14:3112–3120 Ulich TR, Yi ES, Longmuir K et al (1994) Keratinocyte growth
Scott C, Suh J, Stea B et al (2007) Improved survival, quality of factor is a growth factor for type II pneumocytes in vivo.
life, and quality-adjusted survival in breast cancer patients J Clin Invest 93:1298–1306
treated with efaproxiral (Efaproxyn) plus whole-brain Utley JF, Seaver N, Newton GL et al (1984) Pharmacokinetics
radiation therapy for brain metastases. Am J Clin Oncol of WR-1065 in mouse tissue following treatment with WR-
6:580–587 2721. Int J Radiat Oncol Biol Phys 10:1525–1528
Senzer N (2002) A phase III randomized evaluation of Viani GA, Manta GB, Fonseca EC et al (2009) Whole brain
amifostine in stage IIIA/IIIB non-small cell lung cancer radiotherapy with radiosensitizer for brain metastases. J Exp
patients receiving concurrent carboplatin, paclitaxel, and Clin Cancer Res 28:1
radiation therapy followed by gemcitabine and cisplatin Vujaskovic Z, Feng Q, Rabbani ZN et al (2002a) Assessment of
intensification: preliminary findings. Semin Oncol 29:38–41 the protective effect of amifostine on radiation-induced
Shaw LM, Bonner H, Lieberman R (1999) Pharmacokinetic pulmonary toxicity. Exp Lung Res 28:577–590
profile of amifostine. Semin Oncol 23:18–22 Vujaskovic Z, Feng Q, Rabbani ZN et al (2002b) Radiopro-
Shaw E, Scott C, Suh J et al (2003) RSR13 plus cranial tection of lungs by amifostine is associated with reduction
radiation therapy in patients with brain metastases: com- in profibrogenic cytokine activity. Radiat Res 157:
parison with the radiation therapy oncology group recursive 656–660
partitioning analysis brain metastases database. J Clin Oncol Wang LW, Fu XL, Clough R et al (2000) Can angiotension-
21:2364–2371 converting enzyme inhibitors protect against symptomatic
Shepherd FA, Rodrigues Pereira J, Ciuleanu T et al (2005) radiation pneumonitis? Radiat Res 153:405–410
Erlotinib in previously treated non-small-cell lung cancer. Wang R, Kagan R, Tome M (2004) Subcutaneous amifostine
N Engl J Med 353:123–132 during radiation or chemoradiation for the treatment of head
Socinski MA, Morris DE, Halle JS et al (2004) Induction and and neck cancers. ASCO Annual Meeting Proceedings.
concurrent chemotherapy with high-dose thoracic confor- J Clin Oncol 22(14S):8154
mal radiation therapy in unresectable stage IIIA and IIIB Ward HE, Kemsley L, Davies L et al (1992a) The effect of
non-small-cell lung cancer: a dose-escalation phase-I trial. steroids on radiation-induced lung disease in the rat. Radiat
J Clin Oncol 22:4341–4350 Res 136:22–28
Steinauer KK, Gibbs I, Ning S et al (2000) Radiation induces Ward WF, Kim YT, Molteni A et al (1988) Radiation-induced
upregulation of cyclooxygenase-2 protein in PC-3 cells. Int pulmonary endothelial dysfunction in rats: modification by
J Radiat Oncol Biol Phys 48:325–328 an inhibitor of angiotensin converting enzyme. Int J Radiat
Suh JH (2004) Efaproxiral: a novel radiation sensitiser. Expert Oncol Biol Phys 15:135–140
Opin Investig Drugs 13:543–550 Ward WF, Kim YT, Molteni A et al (1992b) Pentoxifylline
Suh JH, Stea B, Nabid A et al (2006) Phase III study of does not spare acute radiation reactions in rat lung and skin.
efaproxiral as an adjunct to whole-brain radiation therapy Radiat Res 129:107–111
for brain metastases. J Clin Oncol 24(1):106–114 Ward WF, Lin PP, Wong PS et al (1993) Radiation
Sumitomo M, Asano T, Asakuma J et al (2004) ZD1839 pneumonitis in rats and its modification by the angioten-
modulates paclitaxel response in renal cancer by blocking sion-converting enzyme inhibitor captopril evaluated by
paclitaxel-induced activation of the epidermal growth factor high resolution computer tomography. Radiat Res 135:
receptor-extracellular signal-regulated kinase pathway. Clin 81–87
Cancer Res 10:794–801 Ward WF, Molteni A, Ts’ao C et al (1990a) Captopril reduces
Tee PG, Travis EL (1995) Basic fibroblast growth factor does collagen and mast cell accumulation in irradiated rat lung.
not protect against classical radiation pneumonitis in two Int J Radiat Oncol Biol Phys 19:1405–1409
strains of mice. Cancer Res 55:298–302 Ward WF, Molteni A, Ts’ao C et al (1990b) The effect of
Terry NHA, Brinkely J, Doig AJ et al (2004) Cellular kinetics captopril on benign and malignant reactions in irradiated rat
of murine lung: model system to determine basis for skin. Br J Radiol 63:349–354
radioprotection with keratinocyte growth factor. Int J Radiat Ward WF, Molteni A, Ts’ao C et al (1992c) Radiation
Oncol Biol Phys 58:435–444 pneumotoxicity in rats: modification by inhibitors of
Radioprotectors and Chemoprotectors in the Management of Lung Cancer 245

angiotensin converting enzyme. Int J Radiat Oncol Biol Yarom R, Harper IS, Wynchangk, et al (1993) Effect of
Phys 22:623–625 captopril on changes in rat’s hearts induced by long-term
Wasserman TH, Phillips TL, Ross G et al (1981) Differential irradiation. Radiat Res 133:187–197
protection against cytotoxic chemotherapeutic effects on Yi ES, Williams ST, Lee H et al (1996) Keratinocyte growth
bone marrow CFUs by WR-2721. Cancer Clin Trials factor ameliorates radiation- and bleomycin-induced lung
4:3–6 injury and mortality. Am J Pathol 149:1963–1970
Weiss JF, Landauer MR (2009) History and development of Yoon S, Park J, Jang H et al (1994) Radioprotective effect of
radiation-protection agents. Int J Radiat Biol 85:539–573 captopril on the mouse jejunal mucosa. Int J Radiat Oncol
Willett CG, Duda DG, Ancukiewicz M et al (2010) A safety Biol Phys 30:873–878
and survival analysis of neoadjuvant bevacizumab with Yuhas JM (1980) Active versus passive absorption kinetics as
standard chemoradiation in a phase I/II study compared the basis for selective protection of normal tissues by S-2-
with standard chemoradiation in locally advanced rectal (3-Aminopropylamino) -ethylphosphorothioic acid. Cancer
cancer. Oncologist 15(8):845–851 Res 40:1519–1524
Systemic Therapy for Lung Cancer
for the Radiation Oncologist
Chandra P. Belani

Contents Abstract
Worldwide, approximately 1.6 million new cases
1 Introduction.............................................................. 248 of lung cancer are diagnosed each year. It contin-
2 Non-Small Cell Lung Cancer................................. 248 ues to be the leading cause of cancer death. With
the use of systemic therapy in addition to radiation
3 Early Stage Resectable Non-Small Cell Lung
Cancer ....................................................................... 249 and surgical resection, the outcome of all lung
cancer patients continues to improve. There is an
4 Systemic Chemotherapy for Early Stage Resected
absolute improvement not only in overall survival
Non-Small Cell Lung Cancer ................................. 249
(OS) but also in the quality of life of these patients,
5 Locally Advanced Unresectable Stage III though modest at best. Non-small cell lung cancer
Disease ....................................................................... 251
(NSCLC) accounts for approximately 87% of all
6 Systemic Therapy for Advanced and Metastatic lung cancers, and is subdivided into two major
Non-Small Cell Lung Cancer ................................. 251
6.1 Histology-based treatment of advanced Non-Small
types, nonsquamous carcinoma and squamous cell
Cell Lung Cancer ...................................................... 252 carcinoma, the former including adenocarcinoma,
6.2 Maintenance therapy ................................................. 252 large cell carcinoma, bronchioloalveolar carci-
6.3 Epidermal Growth Factor Receptor Tyrosine Kinase noma, and poorly differentiated histological sub-
Inhibitors .................................................................... 254
6.4 Mechanisms of Resistance to Epidermal Growth
types. Most patients present after NSCLC have
Factor Receptor ........................................................ 256 spread to regional or distant sites. Patients pre-
senting with advanced, unresectable disease (stage
7 Anti-Angiogenic Therapies for Non-Small Cell
Lung Cancer............................................................. 257 IIIB or IV) and patients who develop recurrent or
metastatic disease following surgical resection are
8 Bevacizumab............................................................. 257
candidates for systemic therapy. Even patients
9 Other Vascular Endothelial Growth Factor with early stage resected disease have improved
Receptor Inhibitors.................................................. 259
OS with the use of adjuvant therapy. There is an
9.1 Other Targeted Approaches for Non-Small Cell
Lung Cancer .............................................................. 259 urgent need to develop novel and effective regi-
mens as the current therapies do not offer a
10 Small Cell Lung Cancer ......................................... 260
curative potential and is palliative with benefits
11 Future Perspectives ................................................. 260 restricted to patients with a good performance
References.......................................................................... 261 status (Eastern Cooperative Oncology Group 0
or 1). The cisplatin-pemetrexed regimen has
C. P. Belani (&) demonstrated preferential activity in patients with
Miriam Beckner Distinguished Professor of Medicine, non-squamous histology. Pemetrexed and erlotinib
Penn State Hershey Cancer Institute,
500 University Drive, CH72, Hershey, have recently emerged as an option for mainte-
PA 17033, USA nance therapy in patients with advanced disease
e-mail: cbelani@psu.edu

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 247
DOI: 10.1007/174_2011_307, Ó Springer-Verlag Berlin Heidelberg 2011
248 C. P. Belani

following four cycles of combination chemother- TRAIL R2, IGF-1R, JAK-2 and the list goes on.
apy leading to a change in treatment paradigm. Thus the major focus of current research has been
Epigenetic alterations have been linked to the the proper selection of patients for optimizing the
pathogenesis and progression of cancer as they effect of molecularly targeted agents with the
lead to the inhibition of transcription of key cell identification and validation of biomarkers.
cycle regulatory genes. Vorinostat (SAHA), with
functional effects on histone acetylation attempts
to restore the normal transcription of cell regula-
tory genes. Provocative activity has been noted 1 Introduction
with the combination of chemotherapy and
vorinostat in advanced NSCLC. Thus, HDAC Lung cancer continues to be the leading cause of
inhibitors are a new class of agents that will cancer deaths worldwide (Jemal et al. 2010b).
require extensive evaluation in lung cancer. Approximately, 1.6 million new cases of lung cancer
Bevacizumab, a monoclonal antibody against the are diagnosed each year (Jemal et al. 2010a). The
vascular endothelial growth factor (VEGF), was number of cases continues to increase in many places
the first agent to demonstrate improved survival in around the world. The overall cure rate from lung
combination with chemotherapy (ECOG 4599) in cancer is modest (approximately 17%) because of the
patients with advanced non-squamous NSCLC. advanced stage at diagnosis in the majority of
The increase in toxicity noted with the addition of patients. The role of systemic therapies has expanded
bevacizumab with chemotherapy calls for caution in the last 2 decades leading to modest improvements
in selecting patients for therapy. The VEGF in overall outcome.
tyrosine kinase inhibitors (TKIs) appear promising,
though they have unique toxicities such as hand–
foot syndrome and fatigue in addition to hyper- 2 Non-Small Cell Lung Cancer
tension. It remains to be seen whether the combi-
nation of VEGF TKI and chemotherapy will Non-small cell lung cancer (NSCLC), which
surpass the efficacy seen with bevacizumab-che- accounts for approximately 87% of all cases of lung
motherapy regimens. Identification or predictive cancer, is a biologically heterogeneous disease. It
biomarkers for patient selection remains elusive includes various histological subtypes such as ade-
for anti-angiogenic agents. Molecular markers for nocarcinoma, bronchioloalveolar carcinoma, squa-
treatment selection of patients with NSCLC are mous cell, and large cell carcinoma. In approximately
increasingly being utilized to personalize therapy. 20–30% of the patients, a specific histological subtype
The presence of an activating mutation in the is not identified, primarily related to lack of adequate
epidermal growth factor receptor (EGFR) is asso- tissue from the diagnostic specimen. Presentation
ciated with high response rates and improved PFS with advanced stage of disease is one of the major
with EGFR TKIs. This has already led to the use of reasons behind the poor overall outcomes for patients
an EGFR TKI as first-line therapy in patients with with NSCLC. In recent years, a number of important
a sensitive EGFR mutation. For patients with wild- advances in therapy have led to improved survival for
type EGFR (or if the EGFR status is unknown) patients with all stages of NSCLC. Combination
chemotherapy remains as the ‘standard of care’ for chemotherapy with platinum-based regimens has led
first-line therapy of advanced NSCLC. The second to improvements in overall survival (OS) and quality
generation EGFR inhibitors are currently under of life in advanced stage disease (Non-small Cell
evaluation. The studies to evaluate the definitive LUng Cancer Collaborative Group 1995; NSCLC
role of an ALK inhibitor in patients with a Meta-Analyses Collaborative Group 2008). For
translocation in the EML 4- ALK gene are in patients with surgically unresectable, locally
progress. Other markers of interest in NSCLC advanced disease, combination of chemotherapy with
include: Excision repair cross-complementing external beam radiation is superior to single modality
gene 1 (ERCC1), RRM1, thymidylate synthase therapy. Even for patients with early stage disease,
(TS), K-ras mutation, c-met expression/mutation, administration of chemotherapy following surgery
Systemic Therapy for Lung Cancer for the Radiation Oncologist 249

results in improved OS (Arriagada et al. 2004; Winton


et al. 2005). Given the extensive role of chemotherapy 4 Systemic Chemotherapy for Early
in NSCLC, development of effective systemic treat- Stage Resected Non-Small Cell
ment regimens is an important priority for physicians Lung Cancer
and researchers.
Approximately, 70% of the patients with early stage
resected disease develop recurrence at distant sites.
3 Early Stage Resectable Non-Small Therefore, in addition to surgical resection, there is
Cell Lung Cancer the need for systemic therapy to eradicate micro-
metastatic disease. Platinum-based chemotherapy
Surgical resection is the foundation of treatment has emerged as an effective adjuvant systemic
for patients with early stage resected NSCLC. therapy post surgical resection. The International
Unfortunately, only 30% of patients have resectable adjuvant lung trial (IALT) with more than 1,700
disease at the time of presentation and 50% of patients comprising stages I, II, or III NSCLC
these have significant co-morbid illness making (Arriagada et al. 2004) randomized after surgery to
them unfit for definitive surgery. For those who are a cisplatin-based two-drug combination versus
medically unresectable, external beam radiation observation (Winton et al. 2005) (Table 1) dem-
therapy (RT) or stereotactic body radiosurgery onstrated a significant but modest 4% improvement
(SBRT) have emerged as promising therapeutic in 5-year survival rate of 4%. The National cancer
options (Timmerman et al. 2010). A randomized institute of Canada (NCIC) trial compared cisplatin
study of SBRT versus surgical resection in vinorelbine versus observation in patients with
medically unresectable patients is in progress. stages IB and II NSCLC (Winton et al. 2005).
Lobectomy continues to be the gold standard and There was an overall 15% improvement in 5-year
lesser resections e.g., sub-lobar, wedge, segmental survival in the adjuvant chemotherapy group.
procedures are associated with increased local fail- A meta-analysis of all recent trials with adjuvant
ure. Pneumonectomy is associated with increased cisplatin-based chemotherapy demonstrated a 5%
morbidity and mortality. Video-assisted thorascopic improvement in OS (Pignon et al. 2008) leading to
surgery (VATS) is increasingly being utilized for a shift of treatment paradigm.
definitive surgical resection (McKenna 1994, 2005; The role of adjuvant chemotherapy has been lim-
Scott et al. 2010). ited to stages II and III resected NSCLC based on the
Adjuvant RT for patients with resected NSCLC preferential benefit in these subgroups from the
was noted to be detrimental in a meta-analysis of a adjuvant therapy studies. Among those with stage I
large number of clinical trials. It is not recommended disease, there continues to be a controversy and there
in patients who undergo an R0 resection as routine are some data to support its use in those with tumors
care. Patients with margin position disease and those C4 cm in size (Strauss et al. 2008). For patients with
with mediastinal node involvement (PORT Meta- stage IA disease, adjuvant chemotherapy is not usu-
analysis Trialists Group 1998) postoperative RT may ally recommended (Pignon et al. 2008).
improve but in addition, these patients benefit from Cisplatin-based combination therapy is the ‘stan-
adjuvant systemic therapy. For patients with early dard of care’ in the adjuvant setting. Whether carbo-
stage NSCLC that are not candidates for surgery due platin can be substituted for cisplatin in these
to medical co-morbid illness, external beam radiation combination regimens when utilized in the adjuvant
is used to treat the tumor. SBRT has emerged as a setting remains to be a subject of controversy.
highly promising therapeutic option for these patients The Cancer and Leukemia Group B (CALGB) trial
and in one of the phase II studies for medically of carboplatin/paclitaxel combination in patients with
unresectable patients, 97% primary tumor control sets stage 1B disease failed to show a survival benefit,
was seen at 3 years (Timmerman et al. 2010). despite improvement in disease-free survival (Strauss
Currently, there is an ongoing European trial et al. 2008). The optimal number of cycles of adjuvant
comparing SBRT to surgical resection in medically chemotherapy has also not been addressed in
resectable NSCLC. randomized studies. Based on the present evidence,
250 C. P. Belani

Table 1 Adjuvant chemotherapy for early stage resected NSCLC


Adjuvant lung Big lung International JBR 10 National Adjuvant navelbine
project trial(BLT) adjuvant lung Cancer Institute of international trialist
Italy(ALPI) (Stephens trial(IALT) Canada (Winton et al. association (ANITA)
(Scagliotti 2005) et al. 2002) (Arriagada et al. 2005) (Douillard et al. 2006)
2004)
Design MVPa vs. Cisplatin Cisplatin doublet vs. Cisplatin vinorelbine Cisplatin vinorelbine vs.
observation doublet vs. observation vs. observation observation
observation
Median 36.5 vs. 27 vs. – - 36.3 vs. 20.7 months HR
PFS 28.9 months HR 24.7 months 0.8; P = 0.017
0.89; P = 0.128 HR 0.97;
P = 0.81
Median 55.2 vs. 33.9 vs. – 94 vs. 73 months HR 65.7 vs. 43.7 months HR
survival 68 months HR 32.6 months 0.69; P = 0.04 0.8; P = 0.017
0.96; P = 0.589 HR 1.02;
P = 0.9
5-year – – 44.5 vs. 40.4% HR – –
Survival 0.86; P \ 0.03
Comment No benefit No benefit Overall 4.1% Benefit limited to Degree of benefit higher
absolute benefit in stage II patients with stage
survival
a
MVP -Mitomycin, Vindesine, Cisplatin

3–4 cycles of cisplatin-based chemotherapy are alone. The power of this study was limited and there
administered in routine practice settings. was a high proportion of stage I patients who
Approximately two-thirds of all resected patients supposedly do not benefit from systemic therapy.
are able to receive adjuvant chemotherapy as others Neo-adjuvant therapy is an efficacious and safe
have co-morbid illness of varying degree and/or approach for patients with early stage NSCLC but
post-operative complications. Neoadjuvant or induc- the ‘standard of care’ for patients with R0 resection
tion therapy has also been investigated to improve the is adjuvant chemotherapy. Integration of targeted
delivery and compliance of chemotherapy. A phase therapies and proper patient selection will lead to
III study conducted by the Southwest oncology tailor-made therapies with resultant improvement in
group demonstrated an improvement in OS with outcome.
neoadjuvant chemotherapy followed by surgery ver- Excision repair cross-complementing gene 1
sus surgery alone (Pisters et al. 2010). However, (ERCC1) is an important mediator of DNA repair and
these differences did not reach statistical significance has been noted in several studies to be a determinant
and the trial was closed early because adjuvant of sensitivity to platinum-based therapy (Lord et al.
chemotherapy became the new ‘standard of care’. 2002; Simon et al. 2007). In a subset analysis of the
Similar data has also been reported from a European IALT study, tumor specimens of patients were eval-
trial that evaluated pre-operative therapy (Scagliotti uated by immunohistochemistry for ERCC1 expres-
et al. 2008a), neoadjuvant chemotherapy prior to sion (Olaussen et al. 2006). Patients with high ERCC1
surgery versus surgery alone versus surgery followed expression did not appear to derive benefit from cis-
by adjuvant chemotherapy were compared in the platin-based chemotherapy, though the OS for this
Spanish study (Felip et al. 2010). The delivery of group was more favorable. In the ERCC1 negative
chemotherapy was found to be superior in the pre- group, adjuvant chemotherapy was associated with a
operative setting (90 vs. 66%) (Felip et al. 2010). robust survival advantage over observation. This
Neo-adjuvant chemotherapy in this Spanish trial was observation has led to prospective studies that eval-
associated with a non-significant trend towards uate treatment selection based on ERCC1 expression
longer disease-free survival compared to surgery for patients with early stage NSCLC.
Systemic Therapy for Lung Cancer for the Radiation Oncologist 251

In the adjuvant setting, epidermal growth factor prognosis and is a prime consideration in treatment
receptor (EGFR) inhibitors have also been investi- selection. Recently, gender has also become recog-
gated for patients with resected early stage NSCLC. nized as an important prognostic factor, with females
Though gefitinib as adjuvant therapy failed to dem- experiencing a better survival than males (Siddiqui
onstrate a benefit in this group it was not conclusive et al. 2010; Batevik et al. 2005; Harichand-Herdt and
as the study was stopped early (Goss et al. 2010). Ramalingam 2009; Visbal et al. 2004).
Erlotinib has been evaluated in a randomized trial in Elderly patients (age C70 years) account for
the adjuvant setting (RADIANT). The trial has com- approximately 50% of all cases of NSCLC (Langer
pleted accrual and the results are eagerly awaited. A et al. 2002). In contrast, patients with a poor perfor-
combination of bevacizumab with standard chemo- mance status (ECOG 2, 3 or 4) have short median
therapy is the subject of evaluation in the Eastern survival duration of approximately 4 months (Ruck-
Cooperative Oncology Group 1505 trial. deschel et al. 1986).
Systemic therapy remains the mainstay or treat-
ment of advanced stage NSCLC. Combination che-
5 Locally Advanced Unresectable motherapy with a platinum-based regimen has
Stage III Disease emerged as the standard therapy for patients with
advanced stage disease (Bunn 2002). Improvements
Combined modality approaches involving systemic in OS and quality of life have been demonstrated with
therapy and radiotherapy are the standard of care for platinum-based regimens over supportive care alone
patients with locally advanced unresectable disease. in randomized clinical trials (Rapp et al. 1988).
Tumors that invade the mediastinum, major blood Among the platinum compounds, both cisplatin and
vessels, heart, or the vertebral body are not considered carboplatin have been extensively studied for the
surgically amenable, in addition to those with treatment of NSCLC. In general, carboplatin-based
multi-station N2 disease. The addition of systemic regimens have a favorable tolerability profile over
chemotherapy to radiation therapy has been associated cisplatin-based regimens (Schiller et al. 2002; Kelly
with improvement in OS compared to radiotherapy et al. 2001). Despite the marginally higher response
alone in randomized studies (Dillman et al. 1990; Le rate with cisplatin-based regimens, considering the
Chevalier et al. 1991). Though the sequential of che- palliative intent of therapy, carboplatin-based regi-
motherapy followed by RT is superior to definitive RT mens have found wide applicability in routine care.
alone, the concurrent approach has clearly shown to be However, recent improvements in anti-emetic therapy
more efficacious as compared to the sequential have rendered the use of cisplatin-based regimens
approach. The role of chemoradiotherapy controver- more tolerable.
sies and consensus regarding the choice of agents and A number of randomized clinical trials have
combination regimens is described elsewhere in this established the superiority of a platinum-containing
book. Patients with locally advanced unresectable two-drug combination over single agent therapy
disease are in general treated with concurrent or (Wozniak et al. 1998; Sandler et al. 2000; Lilenbaum
sequential chemotherapy and definitive RT. These et al. 2005). The response rate, progression-free
approaches result in 20–25% overall 5-year survival survival, and OS all appear to be improved with
(Curran et al. 2000; Furuse et al. 1999). combination regimens in patients with advanced
NSCLC, though the benefits come with higher
toxicity. Paclitaxel, docetaxel, gemcitabine, vinorel-
6 Systemic Therapy for Advanced bine, irinotecan, and pemetrexed, commonly referred
and Metastatic Non-Small Cell to as the ‘third generation’ cytotoxic agents, have all
Lung Cancer demonstrated efficacy when given in combination
with a platinum compound in patients with advanced
The overall goals of treatment are to improve symp- NSCLC (Schiller et al. 2002; Wozniak et al. 1998;
toms, preserve or improve quality of life, and prolong Ohe et al. 2007; Fossella et al. 2003; Scagliotti et al.
survival. The performance status of the patient 2008b; Belani et al. 2005). A large randomized
remains an important determinant of overall clinical trial conducted by the ECOG compared the
252 C. P. Belani

efficacy of four commonly used combination regi- be due to low level of expression of thymidylate
mens in the treatment of advanced NSCLC (Schiller synthase (TS), a known target for pemetrexed
et al. 2002). Cisplatin–docetaxel, cisplatin–gemcita- (Eismann et al. 2005; Righi et al. 2010) in adeno-
bine, and carboplatin–paclitaxel were all associated carcinoma compared to squamous or small cell
with comparable efficacy parameters to that of the carcinoma (Scagliotti et al. 2009).
control arm of cisplatin–paclitaxel. These observa- Another novel chemotherapeutic agent, albumin-
tions, supported by findings of other contemporane- bound paclitaxel when combined with carboplatin
ous clinical trials, established the notion that an showed improved response rate in patients with
‘efficacy plateau’ had been reached with two-drug advanced NSCLC when compared to the standard
combinations in advanced stage NSCLC. The use of combination of paclitaxel and carboplatin (37 vs.
three-drug cytotoxic combinations has generally 25%) with a signal of even larger benefit (41 vs. 24%)
resulted in higher toxicity without clear evidence of in those with squamous cell subset (Socinski et al.
improvement in efficacy across clinical trials and has 2010). Expression of Secreted Protein Acidic and
therefore not been pursued subsequently (Alberola Rich in Cysteine (SPARC), which facilitates accu-
et al. 2003). With the currently available platinum- mulation of albumin in the tumor and thus increase
based two-drug regimens, the median survival and intracellular concentrations of the nab-paclitaxel may
1-year survival rate are 8–11 months and 30–40% in be the reason for the increased efficacy but needs
patients with a good performance status (Ramalingam validation (Desai et al. 2009). Survival data from the
and Belani 2004). trial is awaited before it can be introduced to the
routine care of NSCLC. Thus for now, histology may
represent a harbinger of activity with specific agents
6.1 Histology-based treatment and regimens though eventually there may be a
of advanced Non-Small Cell Lung plausible relationship with a molecular/correlative/
Cancer biological marker.

Choice of systemic therapy based on the sub-histol-


ogy of NSCLC is a new paradigm. Scagliotti et al. 6.2 Maintenance therapy
(2008b) showed that cisplatin-pemetrexed combi-
nation was associated with increased efficacy in the Until recently, 4–6 cycles of combination chemo-
nonsquamous subset of patients. (Scagliotti et al. therapy formed the ‘standard of care’ for patients with
2008b). Over 1700 patients were randomized to the advanced NSCLC (Socinski et al. 2002; Smith et al.
combination of cisplatin–pemetrexed versus cis- 2001) (Table 2). Extension of the same treatment
platin–gemcitabine. The study met its non-inferiority failed to demonstrate any evidence of benefit. Recent
objective and in all NSCLC patients there was no trials of maintenance therapy in stable/responding
difference in survival between the two groups. patients to front-line regimen have shifted the treat-
The cisplatin–pemetrexed regimen demonstrated ment paradigm in favor of this approach. Pemetrexed
preferential activity in patients with non-squamous and erlotinib administered as single agents are
histology. In patients with adenocarcinoma histology, approved by the FDA and EMZA for maintenance
the median survival with the cisplatin–pemetrexed therapy based on the results of randomized trials
regimen was 12.6 m versus 10.9 with cisplatin– (Ciuleanu et al. 2009; Cappuzzo et al. 2010). The
gemcitabine and was statistically significant. Though agents are fairly well tolerated and are devoid of
the exact reasons behind the histology-interaction and significant cumulative toxicity. Furthermore, benefit
pemetrexed are not known but the outcome was of maintenance therapy is related to the ability to
significantly improved in the adenocarcinoma subset. tolerate an active agent following initial chemother-
In addition, this regimen was also associated with a apy rather than at the time of recurrence/progression
favorable tolerability profile. These results led to the (Table 3) though the outcome of those who receive
approval of the cisplatin–pemetrexed regimen for maintenance versus those who are able to receive
patients with only non-squamous NSCLC. Improved treatment at the time of progression may be the same
efficacy of pemetrexed in adenocarcinoma may in fact (Fidias et al. 2009), approximately 35–40% of those
Systemic Therapy for Lung Cancer for the Radiation Oncologist 253

Table 2 Clinical trials of maintenance therapy in advanced NSCLC: prolonged or continuation maintenance
Smith et al. (2001) Socinski et al. (2010) Brodowicz et al. Park et al. (2007) Belani et al. (2010)
(2006)
Design MVP 9 3 cycles vs. PC 9 4 cycles vs. PC GC vs. GC PC 9 4 cycles vs. GC followed by BSC vs.
MVP 9 6 cycles until progression followed by G PC 9 6 cycles GC followed by G
PFS 5 vs. 5 months – 5 vs. 6.2 vs. 4.3 months 7.4 vs. 7.7 months
6.6 months P = 0.001 P = 0.575
P = 0.0001
OS 6 vs. 7 months 8.5 vs. 6.6 P = 0.63 11 vs. 15 vs. 16 months 8 vs. 9.3 months
13 months P = 0.469 P = 0.838
P = 0.469

Table 3 Clinical trials of switch maintenance in advanced NSCLC


Westeel Fidias et al. (2009) SATURN trial ATLAS trial (Miller et al. IFCT-GFPC0502
et al. (Cappuzzo et al. 2009) (Perol et al. 2010)
(2005) 2010)
Design MIC vs. GC followed Platinum doublet Platinum GC followed by BSC
MIC immediately by D vs. followed by doublet ? Bevacizumab vs. G vs. Erlotinib
followed GC followed by D at placebo vs. followed by: Bevacizumab
by V progression Erlotinib vs. Erlotinib ? Bev
PFS 5 vs. 5.7 vs. 2.7 months 11.1 vs. 4.76 vs. 3.75 P = 0.0012 2.9 vs. 1.9 months
3 months P = 0.001 12.3 weeks erlotinib (P = 0.002)
P = 0.32 P = 0.001 3.8 vs. 1.9–G
(P = 0.001)
OS 10.4 vs. 12.3 vs. 9.7 months 12 vs. 11 months 15.9 vs. 13.9 months –
11 months P = 0.853 P = 0.088 P = 0.90

who have initial response are not able to make it to and in the USA. The meta-analysis of maintenance
true second-line intervention. In the intent-to-treat therapy studies demonstrates a significant improve-
analysis, pemetrexed demonstrated a 5.3 month ment in progression-free survival and a modest
improvement in median survival over placebo main- improvement in OS (Soon et al. 2009).
tenance in the non-squamous patients with NSCLC There is continued controversy among lung cancer
leading to its approval by the FDA. A word of caution care regarding the optimal patient for the maintenance
is that those with a poor PS (C2) are not candidates therapy, the choice of agent (continuation of the same
for maintenance therapy. agent vs. switch to a new agent). For now ‘‘switch
Similarly erlotinib, an EGFR TKI, has also shown maintenance’’ has been established until new data
improvement in PFS and OS in the maintenance set- become available and those patients with poor or
ting in responding and stable patients [progression- declining PS should not be offered maintenance
free survival (12.3 vs. 11.1 weeks) and OS (12 vs. therapy (Belani et al. 2010). Ongoing studies evalu-
11 months)] (Cappuzzo et al. 2010). The benefit is ating maintenance are the paramount trial and the
more provocative with erlotinib in those with acti- POINTBREAK STUDIES (Figs. 1, 2). These will
vating mutations in the EGFR TK domain although it provide further insights into this new treatment par-
is modest at best in the overall populations. Both adigm. Bevacizumab and Cetuximab were continued
Pemetrexed and erlotinib have established the foun- as maintenance therapy in the investigational arms of
dation for the maintenance therapy paradigm and are ECOG 4599 & FLEX trials (Perol et al. 2010) but the
approved by the regulatory agencies both in Europe definite role of these agents in the maintenance setting
254 C. P. Belani

Fig. 1

Fig. 2

has not been established (Sandler et al. 2006; Pirker it to second-line therapy and lack of crossover to the
et al. 2009). Prolonged use of the targeted agent fol- ‘‘agent’’ is the placebo arm at progression will con-
lowing 4–6 cycles of cytotoxic therapy has been tinue to pose questions but it is impossible to develop
adopted as a quasi ‘standard of care’. ECOG 5508 is a design of a trial which would attempt to answer
evaluating the role of continuation bevacizumab these questions.
versus pemetrexed versus the combination versus the
combination of the two in patients who have stable or
responsive disease after initial treatment with carbo- 6.3 Epidermal Growth Factor Receptor
platin, paclitaxel, and bevacizumab. Tyrosine Kinase Inhibitors
Maintenance therapy paradigm is increasingly
being adopted. It is fair to say for now that despite EGFR pathway has been implicated in driving tumor
chemo follow-up of responding and stable patients, growth, proliferation, inhibition of apoptosis, increased
approximately a third of patients are not able to make metastatic potential, and initiation of neo-angiogenesis
Systemic Therapy for Lung Cancer for the Radiation Oncologist 255

Table 4 First-line therapy with EGFR TKIs for advanced NSCLC: Randomized studies in enriched patient populations
Trial Selection Design PFS OS
method
(WJTOG3405a Trial) EGFR Gefitinib vs. 9.2 vs. 6.3 months Not reported
(Mitsudomi et al. mutation cisplatin/ P \ 0.0001
2010) docetaxel
Rosell et al. (2009) EGFR Erlotinib 14.0 months (95% 27.0 months (95% CI, 22.7–31.3)
mutation CI, 11.3-16.7)
Maemondo et al. EGFR Gefitinib vs. 10.8 vs. 5.4 months 30.5 months in the gefitinib group and
(2010) mutation carboplatin/ P \ 0.001 23.6 months in the chemotherapy group
paclitaxel (P = 0.31)
Mok et al. (2009) Clinical Gefitinib vs. 5.7 m vs. 5.8, HR 18.6 vs. 17.3 months HR 0.91
characteristics carboplatin/ 0.74, months
paclitaxel P \ 0.001
First Signal Trial Clinical Gefitinib vs. 6.6 vs. 6.1 months 21.3 vs. 23.3 P = 0.420
(Lee et al. 2009) characteristics cisplatin/ P = 0.044
gemcitabine
a
West Japan Thoracic Oncology Group

(Baselga 2001). EGFR is expressed in more than 40% selection studies evaluated the role of gefitinib and
of NSCLC tumors. EGFR pathway inhibitors gefitinib erlotinib (Paz-Ares et al. 2006) resulting in high
and erlotinib were evaluated in patients with refractory response rates to the extent of 80% in the subsets
NSCLC. There was evidence of single agent activity in validating the predictive potential of EGFR mutation
approximately 10–20% of the (Perez-Soler et al. 2004; for response to EGFR TKIs (Rosell et al. 2009). The
Kris et al. 2003; Fukuoka et al. 2003) with skin rash and results of a recent landmark phase III study conducted
diarrhea as the most common side effects. NCIC-BR21 in Asia confirmed the role of EGFR mutation as the
established the efficacy of erlotinib in patients with main predictor of outcome with EGFR TKIs (Mok et al.
recurrent (second-line) advanced NSCLC (Shepherd 2009) (Table 4). Utilizing a clinical enrichment
et al. 2005). There was a significant improvement in OS strategy (women with adenocarcinoma, and history
and progression-free survival leading to the approval of of no or light cigarette smoking were randomized to
erlotinib with second-line setting in unselected therapy with either gefitinib or a standard first-line
patients. Gefitinib, however, failed to show a difference chemotherapy regimen of carboplatin and paclitaxel.
in OS when compared to placebo (Thatcher et al. 2005) Gefitinib was better tolerated with overall population
but the patient population in the Iressa survival evalu- and resulted in a superior PFS, the primary endpoint.
ation (ISEL) study as they had more aggressive/pro- Sixty percent of the patients in whom the tumors
gressive disease but the subsets of never-smokers/ were analyzed had presence of EGFR mutation in
Asian ethnicity demonstrated a benefit. Clinical char- exons 19 and 21. The response rates of 60–80% and
acteristics for response to EGFR TKIs in the early trials median survival of 24–30 months in this selected
included female sex adenocarcinoma histology, never- population with EGFR mutations are provocative.
smokers, and those with Asian ethnicity (Paez et al. The I-PASS study established in addition that
2004). Search for a molecular reason for efficacy in administration of gefitinib in patients with wild-type
these subsets led to the discovery of EGFR activity EGFR was not warranted and chemotherapy was the
mutations in the tyrosine kinase domain of the receptor preferred treatment. The Korean Study (Lee et al.
responsible for the selective activity with EGFR TKIs. 2009) confirmed these observations. Randomized
Approximately 0–15% of all NSCLC patients were phase II studies in patients with documented EGFR
found to harbor the mutative among the Western pop- mutation (Maemondo et al. 2010; Mitsudomi et al.
ulation of NSCLC patients while the incidence is much 2010) have now shown with gefitinib as compared
higher (*40%) in those with Asian ethnicity (Paez to chemotherapy establishing a new paradigm of
et al. 2004). Prospective randomized and patient treatment in the front-line setting for selected. In
256 C. P. Belani

Table 5 Irreversible EGFR Drug Class Targets Status


TKIs
BIBW 2992 EGFR/HRE2 EGFR and HER2 Phase III
HKI-272 EGFR/HER2 EGFR and HER2 Phase II
EKB-569 EGFR/HER2 EGFR and HER2 Phase II
CI-1033 Pan-ErbB EGFR, HER2, and HER4 Phase II
PF00299804 Pan-ErbB EGFR, HER2, and HER4 Phase II
AV-412/MP-412 EGFR/HER2 EGFR and HER2 Phase I
EGFR epidermal growth factor receptor, TKIs tyrosine kinase inhibitors

addition, thin trials have also established the neces- 6.4 Mechanisms of Resistance
sity for evaluation of all adenocarcinomas for EGFR to Epidermal Growth Factor
mutation. The value of adding chemotherapy and Receptor
EGFR TKIs in the front-line setting in patients with
tumors harboring the EGFR mutation has not been The efficacy of EGFR inhibitors is limited by pri-
established and there is an early indication of no mary and acquired resistance. Primary resistance to
added benefit in a recently reported randomized trial erlotinib and gefitinib has been associated with in-
(Herbst et al. 2004; Giaccone et al. 2004). frame insertion mutations in EGFR exon 20 which
To test whether the combination might be benefi- reduces sensitivity to these TKIs by about 100-fold
cial in selected patients, a recent trial randomized and in rare cases, with the T 790M missense
never or light smokers with advanced NSCLC to mutation also located on exon 20 (Kobayashi et al.
therapy with erlotinib alone or in combination with 2005). Gene amplification of MET, the receptor for
carboplatin and paclitaxel (Janne et al. 2010) There hepatocyte growth factor may provide an alternate
was no difference in the outcomes between the two stimulus for promoting survival of NSCLC cells
groups even in patients with EGFR mutation, thus (Engelman et al. 2007). MET amplification occurs
excluding a role for combination of EGFR TKIs in in 3–7% of patients who are EGFR TKI na. Sec-
combination with chemotherapy. ondary acquired resistance in those who have been
A completely different biological basis appears to treated with EGFR TKIs for a period of time can
be responsible for anti-cancer effects of agents that also be caused by the missense 7790M mutation and
target the external domain of the EGFR. Cetuximab, this emerges during treatment in nearly half the
a chimeric monoclonal antibody against the EGFR, cases. Other secondary mutations such as D761c
has minimal activity when given as monotherapy have also been associated with EGFR TKI resis-
for patients with advanced stage NSCLC tance (Balak et al. 2006). Also, as a result of
(Hanna et al. 2006). However, when given in genetic heterogeneity the clones with activating
combination with platinum-based chemotherapy, a mutation may decrease or disappear during treat-
modest improvement in OS was noted (11.3 vs. ment with gefitinib or erlotinib, resulting in a
10.1 months) over chemotherapy alone (Pirker et al. selective survival of wild-type EGFR clones that are
2009). However with other combination regimes, resistant to these agents (Jiang et al. 2008). MET
cetuximab has failed to demonstrate significant amplification is also known to moderate acquired
improvement in survival (Lynch et al. 2007). resistance in 20% of cases (Herbst et al. 2008).
Because of differential efficacy of cetuximab based Tumors that undergo epithelial to mesenchymal
on K-ras mutation (Khambata-Ford et al. 2010) transition also exhibit an increase in metastatic
status in patients with colon cancer, molecular potential and less reliance on EGFR (Barr et al.
analyzes have been performed in the NSCLC trials 2008). Second generation irreversible EGFR inhib-
(Pirker et al. 2009; Lynch et al. 2007; Mukohara itors have demonstrated anti-cancer activity in
et al. 2005). No definitive biological marker has tumors resistant to reversible TKIs. These include
emerged to define sensitivity to cetuximab including BIBW 2992, PF-299804 and others (Belani 2010;
K-ras mutation. Boyer et al. 2010) (Table 5).
Systemic Therapy for Lung Cancer for the Radiation Oncologist 257

To delay the emergence of resistance involves addition of 15 mg/kg dose of bevacizumab to che-
combination therapy with inhibitors of the C-MET motherapy. No objective responses were noted for
and EGFR being utilized in a randomized phase II patients who received bevacizumab as monotherapy;
study, erlotinib alone or in combination with ARQ- 4 out of 13 patients with squamous cell histology
197, a small molecular C-MET (Schiller et al. 2010) developed fatal or life-threatening hemoptysis,
demonstrated a significant improvement in progres- whereas bleeding was uncommon in other histological
sion-free survival and OS and has led to an ongoing sub-types. This observation led to the exclusion of
phase III definitive trial. patients with squamous cell histology in subsequent
clinical trials of bevacizumab in NSCLC. Other
adverse events noted with bevacizumab-based regi-
7 Anti-Angiogenic Therapies men included hypertension and proteinuria.
for Non-Small Cell Lung Cancer The promising efficacy data from this phase II
study led to a confirmatory phase III study of
Higher microvessel density and vascular endothelial carboplatin and paclitaxel with our without
growth factor (VEGF) concentrations have been bevacizumab for advanced non-squamous NSCLC
linked with an aggressive NSCLC phenotype by (ECOG 4599) (Sandler et al. 2006). The 15 mg/kg
various investigators (Fontanini et al. 2002). This dose of bevacizumab was given in combination
provided the rationale to evaluate a variety of anti- with standard doses of carboplatin and paclitaxel
angiogenic agents for the treatment of NSCLC. for patients in the experimental arm. Following
Continuing the common trend in clinical investiga- 6 cycles of combination therapy, bevacizumab was
tions, efforts to develop anti-angiogenic therapies for continued as monotherapy for responding patients
NSCLC have mainly focused on patients with or those with stable disease. Patients with brain
advanced stage disease. Bevacizumab was the first metastasis, history of hemoptysis, and predominant
agent to demonstrate survival advantage in patients squamous histology were excluded. The study met
with advanced stage NSCLC and is approved for its primary endpoint of OS which was superior for
routine use in the first-line setting for patients with patients treated with bevacizumab (10.3 m, 12.3 m,
metastatic non-squamous NSCLC. A number of P-0.003) (Table 6).
VEGF TKIs have also demonstrated modest anti- The progression-free survival duration was also
cancer activity as monotherapy in advanced NSCLC, improved with bevacizumab (6.2 vs. 4.5 m,
thus leading to randomized clinical trials that have or P \ 0.001). Treatment was tolerated well overall,
are evaluating novel combination regimens (Socinski with a less than 5% incidence of major bleeding
et al. 2008; Blumenschein et al. 2009a). events. There was a numerically higher incidence of
treatment-related deaths with the addition of
bevacizumab (15 vs. 2). This was the first ran-
8 Bevacizumab domized study to document an improvement in OS
with the addition of a targeted agent to chemo-
Bevacizumab is a monoclonal antibody that binds therapy for advanced NSCLC and also formed the
to all isoforms of VEGF and inhibits activation of basis for approval of this regiment by the FDA. A
the receptor (Ramalingam 2007). It was first eval- second trial conducted outside the USA noted some
uated in NSCLC on a randomized phase II study of similarities with the efficacy data seen in ECOG
carboplatin and paclitaxel given with or without 4599, though a survival benefit was not evident
bevacizumab (Johnson et al. 2004). This regimen (Reck et al. 2009). In this study (AVAiL), patients
was administered to patients with previously were randomized to receive cisplatin and gemcita-
untreated advanced stage NSCLC. Bevacizumab bine with either bevacizumab or placebo. Patients in
was administered at a dose of either 7.5 or 15 mg/kg. the bevacizumab arm were randomized to either the
Patients on the chemotherapy alone were crossed over 7.5 mg/kg dose or 15 mg/kg. The eligibility criteria
to receive bevacizumab as monotherapy upon disease were similar to those of ECOG 4599. The primary
progression. The response rate, median progression- endpoint of the study was changed from OS to PFS
free survival and OS were all improved with the after nearly two-thirds of the patients had been
258 C. P. Belani

Table 6 Bevacizumab in combination with chemotherapy for advanced NSCLC


Author Regimen Phase Response rate Median PFS Median survival
(%) (m) (m)
Sandler et al. Carboplatin, paclitaxel ? Bevacizumab III 35 6.2 12.3
(2006)
Carboplatin, paclitaxel 15 4.5 10.3
Reck et al. Cisplatin, gemcitabine ? Bevacizumab III 34 6.7 13.6
(2009) (7.5 mg/kg)
Cisplatin, gemcitabine ? Bevacizumab 30 6.5 13.4
(15 mg/kg)
Cisplatin, gemcitabine 20 6.1 13.1

enrolled. Neither the toxicity nor the efficacy a trend towards higher treatment-related deaths in
parameters were significantly different between the patients C70 years of age (Ramalingam et al. 2008).
two dose levels of bevacizumab. The median PFS This calls for careful evaluation of patient perfor-
was improved with the addition of bevacizumab, mance status, co-morbid illness, and baseline risk
though the differences were relatively modest factors for toxicities for elderly patients, before
(6.7 vs. 6.1 m, P = 0.003) for the low dose bev- initiation of bevacizumab-based regimens. Another
acizumab and 6.5 versus 6.1 m, P = 0.03 for high setting to exercise caution involves the use of bev-
dose bevacizumab). The median survival was acizumab with thoracic radiotherapy, since tracheal-
approximately 13 months in all three arms of the esophageal fistula formation has been noted in some
study (Table 6). patients (Spigel et al. 2009a). Therefore, its use
Even though patients with brain metastasis were should be avoided in patients receiving concurrent
not included in both these phase III studies, sub- radiotherapy or in the immediate aftermath of thoracic
sequent non-randomized studies have documented the radiation outside the setting of carefully controlled
safety of bevacizumab-based regimens in patients clinical trials.
with treated brain metastasis (Socinski et al. 2009). The documented ability of bevacizumab to
The AVAiL study also noted no increase in incidence enhance the efficacy of chemotherapy has prompted
of bleeding when bevacizumab-based regimen was an US intergroup study to evaluate its role in the
given to patients on full dose anti-coagulation ther- setting of adjuvant chemotherapy (ECOG 1505). This
apy. The safety of bevacizumab has also been docu- ongoing study randomized patients with stage IB, II,
mented when used in combination with other or IIIA NSCLC to treatment with 4 cycles of cis-
commonly used platinum-based doublets used for the platin-based chemotherapy with or without concur-
treatment of advanced NSCLC (Patel et al. 2009; Kris rent bevacizumab.
et al. 2009). Notably, when given in combination with Despite promising phase II data, the combination
carboplatin and pemetrexed, bevacizumab was asso- of bevacizumab with erlotinib, an inhibitor of the
ciated with a median survival of approximately EGFR, failed to improve survival in a randomized
14 months and a median PFS of 7.8 months (Patel study conducted for second-line therapy of advanced
et al. 2009). Taken together, bevacizumab can be used stage NSCLC (Herbst et al. 2007; Hainsworth 2008).
safely for the treatment of patients with advanced The same combination used as maintenance
non-squamous NSCLC and leads to modest therapy also failed to improve survival compared to
improvements in efficacy. bevacizumab alone (Miller et al. 2009). Thus for
The use of bevacizumab has also met with some patients who receive chemotherapy in combination
challenges in certain patients subsets. An unplanned with bevacizumab as first-line therapy, bevacizumab
subset analysis of the ECOG 4599 study noted a is used as maintenance monotherapy, though its
higher incidence of certain adverse events such as utility has not been confirmed in a randomized
fever with neutropenia, hypertension, proteinuria, and clinical trial.
Systemic Therapy for Lung Cancer for the Radiation Oncologist 259

vandetanib is an active agent in the treatment of


9 Other Vascular Endothelial Growth advanced NSCLC. However, given the relatively
Factor Receptor Inhibitors modest efficacy, it is unlikely to be useful for routine
clinical care in any of the settings mentioned here. An
A number of novel multi-kinase inhibitors which also ongoing phase III study compares vandetanib to pla-
target the VEGF receptor have all been tested for the cebo for patients who progressed following treatment
treatment of advanced NSCLC. Sorafenib, Sunitinib, with erlotinib for advanced NSCLC.
Axitinib, Vandetanib, Vatalanib and Linifanib Cediranib, a potent inhibitor of VEGF-R2, was
(Socinski et al. 2008; Blumenschein et al. 2009a; combined with carboplatin and paclitaxel for first-line
Schiller et al. 2009; Natale et al. 2009; Gauler et al. therapy of advanced NSCLC by the NCIC (Laurie
2007; Tan et al. 2009) have demonstrated evidence of et al. 2008). This randomized phase II study noted
single agent activity and anticancer effects in NSCLC. favorable efficacy trends with the addition of cedira-
When given in combination with chemotherapy, nib, though the toxicity profile has warranted a
Sorafenib failed to show an improvement in survival reduction in the dose of cediranib to 20 mg/day for
(Hanna et al. 2008) and in fact patients with squamous the subsequent follow-up study. Axitinib is another
cell histology, the placebo group fared better. When well tolerated agent that had demonstrated robust
combined with erlotinib in the second-line recurrent single agent activity and is currently being developed
NSCLC, sorafenib demonstrated a modest improve- in combination with chemotherapy (Schiller et al.
ment in efficacy in unselected patients when compared 2009). Linifanib (ABT-869) is a potent VEGF-R2
to erlotinib alone (PFS 1.9 vs. 3.1 m and OS 6 vs. inhibitor that has also demonstrated single agent
8.1 m) (Spigel et al. 2009b). The results of a recently activity in advanced NSCLC and is currently being
completed study of sorafenib versus placebo in heav- tested in a randomized phase II study with combina-
ily treated patients are eagerly awaited. A phase II tion chemotherapy (Tan et al. 2009). The combination
study evaluating sunitinib in combination with of motesanib, a VEGF receptor inhibitor, with
erlotinib versus erlotinib and placebo is in progress. carboplatin and paclitaxel was compared to the same
Sunitinib is also being studied in the maintenance chemotherapy doublet with bevacizumab in a ran-
following four cycles of combination chemotherapy domized phase II study (Blumenschein et al. 2009b).
(CALGB study). Vandetanib is a dual inhibitor of the The two treatment arms were associated with com-
EGFR and VEGF receptor tyrosine kinases. It has also parable efficacy though there was some distinct
been studied in various settings for the treatment of additional toxicity with the oral agent. All of these
advanced stage NSCLC. In the front-line treatment of agents have varying inhibitory concentrations, bio-
advanced NSCLC, the combination of carboplatin and availability, toxicity profile, and anti-cancer activity.
paclitaxel with vandetanib was associated with a It is hoped that one or more of these agents may
modest improvement in median PFS over that of emerge as efficacious in the near future.
the same chemotherapy given without vandetanib
(Heymach et al. 2008). In the second-line setting,
two phase III studies have been completed with 9.1 Other Targeted Approaches
vandetanib. In the first study, docetaxel was given for Non-Small Cell Lung Cancer
alone or in combination with vandetanib (100 mg/day
dose) (Herbst et al. 2009). There was a modest and Echinoderm microtubule-associated protein-like 4
significant improvement in median PFS, though OS gene and the anaplastic lymphoma kinase fusion gene
was not improved. In the second study, vandetanib (EML4-ALK) has been identified in 4–5% of patients
was added to pemetrexed for second-line therapy (De with NSCLC (Soda et al. 2007). ALK translocations
Boer et al. 2009). Though a numerical improvement in occur in young never-smokers with adenocarcinoma
PFS was noted, the differences did not reach statistical (Shaw et al. 2009). NSCLC patients with ALK
significance. Vandetanib was also compared directly translocation appear to have reduced sensitivity to
to erlotinib in a phase III study for advanced NSCLC both standard chemotherapy and EGFR TKIs.
ans was noted to have comparable efficacy (Natale Crizotinib (PF-234066), a small molecule TKI is
et al. 2009). Taken together, these results suggest that known to inhibit ALK kinase. In a single arm study of
260 C. P. Belani

82 patients with ALK translocation or presence of Recently, targeted agents such as anti-angiogenic
EML4-ALK fusion protein (Bang et al. 2010), a agents, mTOR inhibitors, and Bcl-2 inhibitors have
provocative response was noted with crizotinib. The been studied without much success (Pandya et al.
disease control rate of 87% was noted. A Phase II 2007; Rudin et al. 2008). Topotecan, a topoisomerase
study comparing crizotinib to standard chemotherapy inhibitor is the only proven agent in patients that
(docetaxel or pemetrexed) in patients w with EML4 relapsed following platinum-based chemotherapy,
ALK-positive second-line patients with recurrent though its efficacy is restricted to patients with che-
NSCLC is ongoing. Screening for EML4-ALK motherapy-sensitive disease (von Pawel et al. 1999).
translocation by fluorescence in situ hybridization Higher response rate than that with topotecan has
(FISH) technique in never/light smokers with adeno- been noted with amrubicin, an anthracycline deriva-
carcinoma is increasingly being utilized in the clinical tive, in phase II studies for refractory SCLC. This is
setting. This is one successful example of a targeted now being evaluated in a phase III study for second-
approached in NSCLC. line therapy (Onoda et al. 2006).
Other targeted agents in development for NSCLC The use of thoracic radiotherapy and prophylactic
include but are not limited to HDAC (histone deace- cranial irradiation (PCI) are associated with modest
tylase) inhibitors (Owonikoko et al. 2010; Ramalingam improvements in survival for patients with limited-
et al. 2010a), mTOR inhibitors (Sun et al. 2006; Hudes stage disease. Recently, a randomized study in
et al. 2007), IGF-IR inhibitors (Ramalingam et al. patients with extensive stage SCLC demonstrated an
2010b; Soria et al. 2009). improvement in OS with PCI. Though this study was
limited in not staging the brain before initiation of
PCI, the robust survival results have led to the con-
10 Small Cell Lung Cancer sideration of PCI in patients who experience a good
response to combination chemotherapy.
SCLC cases account for only 13% of all lung cancers Ongoing studies are evaluating newer classes of
in the Western world and the numbers continue to molecularly targeted agents such as the Hedgehog
shrink (Govindan et al. 2006). Despite a reduction in inhibitors and the IGF-1R pathway inhibitors.
the percentage of patients with SCLC in recent times, A greater understanding of mediators of resistance
it is still a considerable source of morbidity and and sensitivity to platinum is also necessary to
mortality with more than 25,000 new cases diagnosed improve the efficacy of combination chemotherapy
each year in the USA (Govindan et al. 2006). With the for SCLC.
adoption of the new staging system, the use of the
TNM staging has been recommended over the pre-
vailing categorization of SCLC into limited and 11 Future Perspectives
extensive stage disease (Vallieres et al. 2009). This
will allow for an improvement in the ability to The treatment options for patients with lung cancer
establish prognosis. have improved considerably in recent years with
Platinum-based chemotherapy continues to be the modest benefit in outcome measures. Adjuvant che-
cornerstone of treatment for SCLC and very little motherapy for early stage resected disease and
progress has been achieved in the past three dec- maintenance therapy for advanced stage response
ades. Four cycles of combination therapy with a disease have emerged as new treatment paradigms in
platinum compound and etoposide remains the the last decade. The success with selective use of
‘standard of care’ for both limited (with thoracic EGFR TKIs in a targeted population has led to per-
radiotherapy) and extensive stage SCLC. Irinotecan, sonalized approaches to treatment. Deciphering of the
a topoisomerase inhibitor failed to show a survival dominant oncogenic drivers in the tumors from
advantage (seen in the Japanese study) (Noda et al. patients with lung cancer and increase in the know-
2002) when given in combination with cisplatin and ledge of lung cancer biology will be of utmost
compared to the gold standard of cisplatin– importance in guiding various treatment methods and
etoposide. strategies.
Systemic Therapy for Lung Cancer for the Radiation Oncologist 261

non-squamous non-small cell lung cancer. J Thorac Oncol


References 4(9):S354 (Abs# C1.4)
Boyer MJ, Blackhall FH, Park K, Barrios C, Krzakowski M,
Taylor I et al (2010) Efficacy and safety of PF299804
Alberola V, Camps C, Provencio M, Isla D, Rosell R, Vadell C versus erlotinib (E): aglobal, randomized phase II trial in
et al (2003) Cisplatin plus gemcitabine versus a cisplatin- patients (pts) with advanced non-small cell lung cancer
based triplet versus nonplatinum sequential doublets in (NSCLC) after failure of chemotherapy (CT). J Clin Oncol
advanced non-small-cell lung cancer: a Spanish Lung 28(15s)
Cancer Group phase III randomized trial. J Clin Oncol Brodowicz T, Krzakowski M, Zwitter M, Tzekova V, Ramlau R,
21(17):3207–3213 Ghilezan N et al (2006) Cisplatin and gemcitabine first-line
Arriagada R, Bergman B, Dunant A, Le Chevalier T, Pignon JP, chemotherapy followed by maintenance gemcitabine or best
Vansteenkiste J (2004) Cisplatin-based adjuvant chemo- supportive care in advanced non-small cell lung cancer: a
therapy in patients with completely resected non-small-cell phase III trial. Lung Cancer 52(2):155–163
lung cancer. N Engl J Med 350(4):351–360 Bunn PA Jr (2002) Chemotherapy for advanced non-small-cell
Balak MN, Gong Y, Riely GJ, Somwar R, Li AR, Zakowski MF lung cancer: Who, what, when, why? J Clin Oncol 20(18
et al (2006) Novel D761Y and common secondary T790 M Suppl):23S–33S
mutations in epidermal growth factor receptor-mutant lung Cappuzzo F, Ciuleanu T, Stelmakh L, Cicenas S, Szczesna A,
adenocarcinomas with acquired resistance to kinase inhib- Juhasz E et al (2010) Erlotinib as maintenance treatment in
itors. Clin Cancer Res 12(21):6494–6501 advanced non-small-cell lung cancer: a multicentre, ran-
Bang Y, Kwak EL, Shaw A, Camidge R, Lafrate J, Maki R et al domised, placebo-controlled phase 3 study. Lancet Oncol
(2010) Clinical activity of the oral ALK inhibitor PF- 11(6):521–529
02341066 in ALK-positive patients with non-small cell lung Ciuleanu T, Brodowicz T, Zielinski C, Kim JH, Krzakowski M,
cancer (NSCLC). J Clin Oncol 28(15s) (Abs# 3) Laack E et al (2009) Maintenance pemetrexed plus best
Barr S, Thomson S, Buck E, Russo S, Petti F, Sujka-Kwok I supportive care versus placebo plus best supportive care for
et al (2008) Bypassing cellular EGF receptor dependence non-small-cell lung cancer: a randomised, double-blind,
through epithelial-to-mesenchymal-like transitions. Clin phase 3 study. Lancet 374(9699):1432–1440
Exp Metastasis 25(6):685–693 Curran W, Scott C, Langer CJ, Komaki R, Lee JS, Hauser S
Baselga J (2001) The EGFR as a target for anticancer therapy— et al (2000) Phase III comparison of sequential vs concur-
focus on cetuximab. Eur J Cancer 37(Suppl 4):S16–S22 rent chemoradiation for patients (Pts) with unresected stage
Batevik R, Grong K, Segadal L, Stangeland L (2005) The III non-small cell lung cancer (NSCLC): Initial Report of
female gender has a positive effect on survival independent Radiation therapy Oncology Group (RTOG) 9410. Proc Am
of background life expectancy following surgical resection Soc Clin Oncol 19:484a
of primary non-small cell lung cancer: a study of absolute De Boer R, Arrieta Ó, Gottfried M, Blackhall F, Raats J,
and relative survival over 15 years. Lung Cancer Yang C, Langmuir P (2009) Vandetanib plus pemetrexed
47(2):173–181 versus pemetrexed as second-line therapy in patients with
Belani CP (2010) The role of irreversible EGFR inhibitors in advanced non-small cell lung cancer (NSCLC): A random-
the treatment of non-small cell lung cancer: overcoming ized, double-blind phase III trial (ZEAL). J Clin Oncol
resistance to reversible EGFR inhibitors. Cancer Invest 27(15S) (Abs# 8010)
28(4):413–423 Desai N, Trieu V, Damascelli B, Soon-Shiong P (2009) SPARC
Belani CP, Waterhouse DM, Ghazal H, Ramalingam SS, expression correlates with tumor response to albumin-bound
Bordoni R, Greenberg R et al (2010) Phase III study of paclitaxel in head and neck cancer patients. Transl Oncol
maintenance gemcitabine (G) and best supportive care 2(2):59–64
(BSC) versus BSC, following standard combination therapy Dillman RO, Seagren SL, Propert KJ, Guerra J, Eaton WL,
with gemcitabine-carboplatin (G-Cb) for patients with Perry MC et al (1990) A randomized trial of induction
advanced non-small cell lung cancer (NSCLC). J Clin chemotherapy plus high-dose radiation versus radiation
Oncol 28(15s):7506–7506 alone in stage III non-small-cell lung cancer. N Engl J Med
Belani CP, Lee JS, Socinski MA, Robert F, Waterhouse D, 323(14):940–945
Rowland K et al (2005) Randomized phase III trial Douillard JY, Rosell R, De Lena M, Carpagnano F, Ramlau R,
comparing cisplatin-etoposide to carboplatin–paclitaxel in Gonzales-Larriba JL et al (2006) Adjuvant vinorelbine plus
advanced or metastatic non-small cell lung cancer. Ann cisplatin versus observation in patients with completely
Oncol 16(7):1069–1075 resected stage IB-IIIA non-small-cell lung cancer (Adjuvant
Blumenschein GR Jr, Gatzemeier U, Fossella F, Stewart DJ, Navelbine International Trialist Association [ANITA]): a
Cupit L, Cihon F et al (2009a) Phase II, multicenter, randomised controlled trial. Lancet Oncol 7(9):719–727
uncontrolled trial of single-agent sorafenib in patients with Eismann U, Oberschmidt O, Ehnert M, Fleeth J, Ludtke FE,
relapsed or refractory, advanced non-small-cell lung cancer. Struck S et al (2005) Pemetrexed: mRNA expression of the
J Clin Oncol 27(26):4274–4280 target genes TS, GARFT and DHFR correlates with the in
Blumenschein GKF, Memon H, Mok T, Stephenson J, Beck T, vitro chemosensitivity of human solid tumors. Int J Clin
Lakshmaiah K (2009b) Randomized, open-label phase 2 Pharmacol Ther 43(12):567–569
study of safety and efficacy of motesanib or bevacizumab in Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C,
combination with paclitaxel and carboplatin for advanced Park JO et al (2007) MET amplification leads to gefitinib
262 C. P. Belani

resistance in lung cancer by activating ERBB3 signaling. advanced non-small cell lung cncer after failure of standard
Sci 316(5827):1039–1043 first-line chemotherapy (BETA). J Thorac Oncol 3(suppl
Felip E, Rosell R, Maestre JA, Rodriguez-Paniagua JM, Moran T, 4):S302
Astudillo J et al (2010) Preoperative chemotherapy plus Hanna N, Lilenbaum R, Ansari R, Lynch T, Govindan R,
surgery versus surgery plus adjuvant chemotherapy versus Janne PA et al (2006) Phase II trial of cetuximab in patients
surgery alone in early-stage non-small-cell lung cancer. J Clin with previously treated non-small-cell lung cancer. J Clin
Oncol 28(19):3138–3145 Oncol 24(33):5253–5258
Fidias PM, Dakhil SR, Lyss AP, Loesch DM, Waterhouse Dm, Hanna NVPJ, Reck M, Scagliotti G (2008) Carboplatin/
Bromund JL, Chen R, Kazmierski MH, Treat J, Obasaju CK, paclitaxel with/without sorafenib in chemonaive patients
Marciniak M, Gill J, Schiller JH (2009) Phase III study of with stage IIIB-IV non-small cell lung cancer: intermim
immediate compared with delayed docetaxel after front-Line analysis from a randomized phase III trials ESCAPE.
therapy with gemcitabine plus carboplatin in advanced non- J Thorac Oncol 3(11):S268
small-cell lung cancer. J Clin Oncol 27(4):591–598 Harichand-Herdt S, Ramalingam SS (2009) Gender-associated
Fontanini G, Faviana P, Lucchi M, Boldrini L, Mussi A, differences in lung cancer: clinical characteristics and
Camacci T et al (2002) A high vascular count and treatment outcomes in women. Semin Oncol 36(6):572–580
overexpression of vascular endothelial growth factor are Herbst R, Sun Y, Korfee S, Germonpre N, Saijo N, Zhou C,
associated with unfavourable prognosis in operated small Wang J (2009) Vandetanib plus docetaxel versus docetaxel
cell lung carcinoma. Br J Cancer 86(4):558–563 as second-line treatment for patients with advanced non-
Fossella F, Pereira JR, von Pawel J, Pluzanska A, Gorbounova V, small cell lung cancer (NSCLC): a randomized, double-
Kaukel E et al (2003) Randomized, multinational, phase III blind phase III trial (ZODIAC). J Clin Oncol 27(15S) (Abs#
study of docetaxel plus platinum combinations versus 8003)
vinorelbine plus cisplatin for advanced non-small-cell lung Herbst RS, Giaccone G, Schiller JH, Natale RB, Miller V,
cancer: the TAX 326 study group. J Clin Oncol 21(16): Manegold C et al (2004) Gefitinib in combination with
3016–3024 paclitaxel and carboplatin in advanced non-small-cell lung
Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, cancer: a phase III trial-INTACT 2. J Clin Oncol 22(5):785–
Douillard JY et al (2003) Multi-institutional randomized 794
phase II trial of gefitinib for previously treated patients with Herbst RS, O’Neill VJ, Fehrenbacher L, Belani CP, Bonomi PD,
advanced non-small-cell lung cancer (The IDEAL 1 Trial) Hart L et al (2007) Phase II study of efficacy and safety of
[corrected]. J Clin Oncol 21(12):2237–2246 bevacizumab in combination with chemotherapy or erloti-
Furuse K, Fukuoka M, Kawahara M, Nishikawa H, Takada Y, nib compared with chemotherapy alone for treatment of
Kudoh S et al (1999) Phase III study of concurrent versus recurrent or refractory non small-cell lung cancer. J Clin
sequential thoracic radiotherapy in combination with mito- Oncol 25(30):4743–4750
mycin, vindesine, and cisplatin in unresectable stage III Herbst RS, Heymach JV, Lippman SM (2008) Lung cancer.
non-small-cell lung cancer. J Clin Oncol 17(9):2692–2699 N Engl J Med 359(13):1367–1380
Gauler T, Besse B, Meric J, Gounant V, Fischer B, Overbeck T, Heymach JV, Paz-Ares L, De Braud F, Sebastian M, Stewart DJ,
Krissel H (2007) Phase II open-label study to investigate Eberhardt WE et al (2008) Randomized phase II study of
efficacy and safety of TPK787/ZK 222584 (PTK/ZK) orally vandetanib alone or with paclitaxel and carboplatin as first-
administered once daily or twice daily at 1,250 mg as line treatment for advanced non-small-cell lung cancer. J Clin
second-line monotherapy in patients (pts) with stage IIIB/IV Oncol 26(33):5407–5415
non-small cell lung cancer (NSCLC). J Clin Oncol 25(18S) Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A
(Abs# 7541) et al (2007) Temsirolimus, interferon alfa, or both for
Giaccone G, Herbst RS, Manegold C, Scagliotti G, Rosell R, advanced renal-cell carcinoma. N Engl J Med 356(22):
Miller V et al (2004) Gefitinib in combination with 2271–2281
gemcitabine and cisplatin in advanced non-small-cell lung Janne P, Wang XF, Socinski M, Crawford J, Capelletti M,
cancer: a phase III trial-INTACT 1. J Clin Oncol 22(5):777– Edelman M et al (2010) Randomized phase II trial of
784 erlotinib (E) alone or in combination iwth carboplatin/
Goss G, Lorimer I, Tsao MS, O’Callaghan CJ, Ding K, Masters G paclitaxel (CP) in never or light former smokers with
et al (2010) A phase II randomized, double-blind, placebo- advanced lung adenocarcinoma: CALGB 30406. J Clin
controlled trial of the epidermal growth factor receptor Oncol 28(15s)(Abs# 7503)
inhibitor gefitinib in completely resected stage IB-IIIA Jemal A, Siegel R, Xu J, Ward E (2010a) Cancer statistics. CA
non-small cell lung cancer (NSCLC): NCIC CTG BR.19. Cancer J Clin 60(5):277–300
J Clin Oncol 28(15s) Jemal A, Center MM, DeSantis C, Ward EM (2010b) Global
Govindan R, Page N, Morgensztern D, Read W, Tierney R, patterns of cancer incidence and mortality rates and trends.
Vlahiotis A et al (2006) Changing epidemiology of small- Cancer Epidemiol Biomarkers Prev 19(8):1893–1907
cell lung cancer in the United States over the last 30 years: Jiang SX, Yamashita K, Yamamoto M, Piao CJ, Umezawa A,
analysis of the surveillance, epidemiologic, and end results Saegusa M et al (2008) EGFR genetic heterogeneity of
database. J Clin Oncol 24(28):4539–4544 nonsmall cell lung cancers contributing to acquired gefitinib
Hainsworth J, Herbst R (2008) A phase III, multicenter, resistance. Int J Cancer 123(11):2480–2486
placebo-controled, double-blind, randomized clinical trial to Johnson DH, Fehrenbacher L, Novotny WF, Herbst RS,
evaluate the efficacy of bevacizumab in combination with Nemunaitis JJ, Jablons DM et al (2004) Randomized
erlotinib compared with erlotinib alone for treatment of phase II trial comparing bevacizumab plus carboplatin
Systemic Therapy for Lung Cancer for the Radiation Oncologist 263

and paclitaxel with carboplatin and paclitaxel alone in Lynch T, Patel T, Dreisbach L, McCleod M, Robert WJ,
previously untreated locally advanced or metastatic non- Hermann E et al (2007) A randomized phase III study of
small-cell lung cancer. J Clin Oncol 22(11):2184–2191 cetuximab in combination with taxane/carboplatin versus
Kelly K, Crowley J, Bunn PA Jr, Presant CA, Grevstad PK, taxane/carboplatin alone as first-line treatment ofr patients
Moinpour CM et al (2001) Randomized phase III trial of with advanced/metastatic non-small cell lung cancer.
paclitaxel plus carboplatin versus vinorelbine plus cisplatin J Thorac Oncol 2(8 (suppl 4)):S340
in the treatment of patients with advanced non-small-cell Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S,
lung cancer: a Southwest Oncology Group trial. J Clin Isobe H et al (2010) Gefitinib or chemotherapy for non-
Oncol 19(13):3210–3218 small-cell lung cancer with mutated EGFR. N Engl J Med
Khambata-Ford S, Harbison CT, Hart LL, Awad M, Xu LA, 362(25):2380–2388
Horak CE et al (2010) Analysis of potential predictive McKenna RJ Jr (1994) Lobectomy by video-assisted thoracic
markers of cetuximab benefit in BMS099, a phase III study surgery with mediastinal node sampling for lung cancer.
of cetuximab and first-line taxane/carboplatin in advanced J Thorac Cardiovasc Surg 107(3):879–881 (discussion
non-small-cell lung cancer. J Clin Oncol 28(6):918–927 81-2)
Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, McKenna RJ Jr (2005) New approaches to the minimally
Meyerson M et al (2005) EGFR mutation and resistance of invasive treatment of lung cancer. Cancer J 11(1):73–76
non-small-cell lung cancer to gefitinib. N Engl J Med Miller V, O’Connor P, Soh C, Kabbinavar F (2009) A
352(8):786–792 randomized, double-blind, placebo-controlled, phase IIIb
Kris MG, Natale RB, Herbst RS, Lynch TJ Jr, Prager D, Belani trial (ATLAS) comparing bevacizumab (B) therapy with or
CP et al (2003) Efficacy of gefitinib, an inhibitor of the without erlotinib (E) after completion of chemotherpay with
epidermal growth factor receptor tyrosine kinase, in symp- B for first-line treatment of locally advanced, recurrent, or
tomatic patients with non-small cell lung cancer: a metastatic non-small cell lung cancer (NSCLC). J Clin
randomized trial. JAMA 290(16):2149–2158 Oncol 27(15S)
Kris MG, Price KA, Rusch VW, Zhao B, Schwartz L, Guo P, Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I,
Tan Y (2009) Phase II trial of induction and adjuvant Tsurutani J et al (2010) Gefitinib versus cisplatin plus
bevacizumab with cisplatin and docetaxel in patients with docetaxel in patients with non-small-cell lung cancer
locally advanced non-small cell lung cancer. J Thorac harbouring mutations of the epidermal growth factor
Oncol 4(9):S372 receptor (WJTOG3405): an open label, randomised phase
Langer CJ, Manola J, Bernardo P, Kugler JW, Bonomi P, Cella 3 trial. Lancet Oncol 11(2):121–128
D et al (2002) Cisplatin-based therapy for elderly patients Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N
with advanced non-small-cell lung cancer: implications of et al (2009) Gefitinib or carboplatin–paclitaxel in pulmon-
Eastern Cooperative Oncology Group 5592, a randomized ary adenocarcinoma. N Engl J Med 361(10):947–957
trial. J Natl Cancer Inst 94(3):173–181 Mukohara T, Engelman JA, Hanna NH, Yeap BY, Kobayashi
Laurie S, Arnold A, Shepherd F, Dediu M, Ciuleanu T, S, Lindeman N et al (2005) Differential effects of gefitinib
Fenton D, Zukin M (2008) Randomized, double-blind phase and cetuximab on non-small-cell lung cancers bearing
II trial of carboplatin, paclitaxel with either daily oral epidermal growth factor receptor mutations. J Natl Cancer
cediranib, an inhibitor of vascular endothelial growth factor Inst 97(16):1185–1194
receptor tyrosine kinases, or placebo, in advanced non-small Natale NB, Thongprasert S, Greco A, Thomas M, Tsai CM,
cell lung cancer: a study of the National Cancer Institute of Sungpaweravong D, Ferry D (2009) Vandetanib versus
Canada Clinical Trials Group. Ann Oncol 19(suppl 8):290 erlotinib in patients with advanced non-small cell lung
(Abs# 231PD) cancer (NSCLC) after failure of at least one prior cytotoxic
Le Chevalier T, Arriagada R, Quoix E, Ruffie P, Martin M, chemotherapy: A randomized, double-blind phase III trial
Tarayre M et al (1991) Radiotherapy alone versus combined (ZEST). J Clin Oncol 27(15S) (Abs# 8009)
chemotherapy and radiotherapy in nonresectable non-small- Noda K, Nishiwaki Y, Kawahara M, Negoro S, Sugiura T,
cell lung cancer: first analysis of a randomized trial in 353 Yokoyama A et al (2002) Irinotecan plus cisplatin com-
patients. J Natl Cancer Inst 83(6):417–423 pared with etoposide plus cisplatin for extensive small-cell
Lee JS, Park K, Kim S, Lee DH, Kim H, Han JH et al (2009) A lung cancer. N Engl J Med 346(2):85–91
randomized phase III study of gefitinib versus standard Non-small Cell LUng Cancer Collaborative Group (1995)
chemotherapy (gemcitabine plus cisplatin) as first-line Chemotherapy in non-small cell lung cancer: a meta-
treatment for never-smokers with advanced or metastatic analysis using updated data on individual patients from 52
adenocarcimoma of the lung. J Thorac Oncol 4(9 (suppl 1)) randomised clinical trials. BMJ 311(7010):899–909
Lilenbaum RC, Herndon JE II, List MA, Desch C, Watson DM, NSCLC Meta-Analyses Collaborative Group (2008) Che-
Miller AA et al (2005) Single-agent versus combination motherapy in addition to supportive care improves survival
chemotherapy in advanced non-small-cell lung cancer: the in advanced non-small-cell lung controlled trials. J Clin
cancer and leukemia group B (study 9730). J Clin Oncol Oncol 26(28):4617–4625
23(1):190–196 Ohe Y, Ohashi Y, Kubota K, Tamura T, Nakagawa K, Negoro
Lord RV, Brabender J, Gandara D, Alberola V, Camps C, S et al (2007) Randomized phase III study of cisplatin plus
Domine M et al (2002) Low ERCC1 expression correlates irinotecan versus carboplatin plus paclitaxel, cisplatin plus
with prolonged survival after cisplatin plus gemcitabine gemcitabine, and cisplatin plus vinorelbine for advanced
chemotherapy in non-small cell lung cancer. Clin Cancer non-small-cell lung cancer: Four-Arm Cooperative Study in
Res 8(7):2286–2291 Japan. Ann Oncol 18(2):317–323
264 C. P. Belani

Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, preoperative paclitaxel and carboplatin in early-stage non-
Haddad V et al (2006) DNA repair by ERCC1 in non-small- small-cell lung cancer: Southwest Oncology Group Trial
cell lung cancer and cisplatin-based adjuvant chemotherapy. S9900, an intergroup, randomized, phase III trial. J Clin
N Engl J Med 355(10):983–991 Oncol 28(11):1843–1849
Onoda S, Masuda N, Seto T, Eguchi K, Takiguchi Y, Isobe H PORT Meta-analysis Trialists Group (1998) Postoperative
et al (2006) Phase II trial of amrubicin for treatment of radiotherapy in non-small-cell lung cancer: systematic
refractory or relapsed small-cell lung cancer: Thoracic review and meta-analysis of individual patient data from
Oncology Research Group Study 0301. J Clin Oncol nine randomised controlled trials. Lancet 352(9124):257–263
24(34):5448–5453 Ramalingam S, Belani CP (2007) Role of bevacizumab for the
Owonikoko TK, Ramalingam SS, Kanterewicz B, Balius TE, treatment of non-small-cell lung cancer. Future Oncol
Belani CP, Hershberger PA (2010) Vorinostat increases 3(2):131–139
carboplatin and paclitaxel activity in non-small-cell lung Ramalingam S, Belani CP (2004) State-of-the-art chemother-
cancer cells. Int J Cancer 126(3):743–755 apy for advanced non-small cell lung cancer. Semin Oncol
Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S et al 31(1 suppl 1):68–74
(2004) EGFR mutations in lung cancer: correlation with Ramalingam SS, Dahlberg SE, Langer CJ, Gray R, Belani CP,
clinical response to gefitinib therapy. Science Brahmer JR et al (2008) Outcomes for elderly, advanced-
304(5676):1497–1500 stage non small-cell lung cancer patients treated with
Pandya KJ, Dahlberg S, Hidalgo M, Cohen RB, Lee MW, bevacizumab in combination with carboplatin and paclit-
Schiller JH et al (2007) A randomized, phase II trial of two axel: analysis of Eastern Cooperative Oncology Group Trial
dose levels of temsirolimus (CCI-779) in patients with 4599. J Clin Oncol 26(1):60–65
extensive-stage small-cell lung cancer who have responding Ramalingam SS, Maitland ML, Frankel P, Argiris AE,
or stable disease after induction chemotherapy: a trial of the Koczywas M, Gitlitz B et al (2010a) Carboplatin and
Eastern Cooperative Oncology Group (E1500). J Thorac Paclitaxel in combination with either vorinostat or placebo
Oncol 2(11):1036–1041 for first-line therapy of advanced non-small-cell lung
Park JO, Kim SW, Ahn JS, Suh C, Lee JS, Jang JS et al (2007) cancer. J Clin Oncol 28(1):56–62
Phase III trial of two versus four additional cycles in Ramalingam SS, Harvey RD, Saba N, Owonikoko TK, Kauh J,
patients who are nonprogressive after two cycles of Shin DM et al (2010b) Phase 1 and pharmacokinetic study
platinum-based chemotherapy in non small-cell lung cancer. of everolimus, a mammalian target of rapamycin inhibitor,
J Clin Oncol 25(33):5233–5239 in combination with docetaxel for recurrent/refractory
Patel JD, Hensing TA, Rademaker A, Hart EM, Blum MG, nonsmall cell lung cancer. Cancer 116(16):3903–3909
Milton DT et al (2009) Phase II study of pemetrexed and Rapp E, Pater JL, Willan A, Cormier Y, Murray N, Evans WK
carboplatin plus bevacizumab with maintenance pemetr- et al (1988) Chemotherapy can prolong survival in patients
exed and bevacizumab as first-line therapy for nonsquamous with advanced non-small-cell lung cancer—report of a
non-small-cell lung cancer. J Clin Oncol 27(20):3284–3289 Canadian multicenter randomized trial. J Clin Oncol
Paz-Ares L, Douillard JY, Koralewski P, Manegold C, Smit EF, 6(4):633–641
Reyes JM et al (2006) Phase III study of gemcitabine and Reck M, von Pawel J, Zatloukal P, Ramlau R, Gorbounova V,
cisplatin with or without aprinocarsen, a protein kinase C- Hirsh V et al (2009) Phase III trial of cisplatin plus
alpha antisense oligonucleotide, in patients with advanced- gemcitabine with either placebo or bevacizumab as first-line
stage non-small-cell lung cancer. J Clin Oncol therapy for nonsquamous non-small-cell lung cancer:
24(9):1428–1434 AVAil. J Clin Oncol 27(8):1227–1234
Perez-Soler R, Chachoua A, Hammond LA, Rowinsky EK, Righi L, Papotti MG, Ceppi P, Bille A, Bacillo E, Molinaro L
Huberman M, Karp D et al (2004) Determinants of tumor et al (2010) Thymidylate synthase but not excision repair
response and survival with erlotinib in patients with non– cross-complementation group 1 tumor expression predicts
small-cell lung cancer. J Clin Oncol 22(16):3238–3247 outcome in patients with malignant pleural mesothelioma
Perol M, Chouaid C, Milleron B, Gervais R, Barlesi F, Westeel treated with pemetrexed-based chemotherapy. J Clin Oncol
V et al (2010) Maintenance with either gemcitabine or 28(9):1534–1539
erlotinib versus observation with predefined second-line Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C
treatment after cisplatin–gemcitabine induction chemother- et al (2009) Screening for epidermal growth factor receptor
apy in advanced NSCLC: IFCT-GFPC 0502 phase III study. mutations in lung cancer. N Engl J Med 361(10):958–967
J Clin Oncol 28(15s) Ruckdeschel JC, Finkelstein DM, Ettinger DS, Creech RH,
Pignon JP, Tribodet H, Scagliotti GV, Douillard JY, Shepherd Mason BA, Joss RA et al (1986) A randomized trial of the
FA, Stephens RJ et al (2008) Lung adjuvant cisplatin four most active regimens for metastatic non-small-cell lung
evaluation: a pooled analysis by the LACE Collaborative cancer. J Clin Oncol 4(1):14–22
Group. J Clin Oncol 26(21):3552–3559 Rudin CM, Salgia R, Wang X, Hodgson LD, Masters GA,
Pirker R, Pereira JR, Szczesna A, von Pawel J, Krzakowski M, Green M et al (2008) Randomized phase II Study of
Ramlau R et al (2009) Cetuximab plus chemotherapy in carboplatin and etoposide with or without the bcl-2
patients with advanced non-small-cell lung cancer (FLEX): antisense oligonucleotide oblimersen for extensive-stage
an open-label randomised phase III trial. Lancet small-cell lung cancer: CALGB 30103. J Clin Oncol
373(9674):1525–1531 26(6):870–876
Pisters KM, Vallieres E, Crowley JJ, Franklin WA, Bunn PA Jr, Sandler AB, Nemunaitis J, Denham C, von Pawel J, Cormier Y,
Ginsberg RJ et al (2010) Surgery with or without Gatzemeier U et al (2000) Phase III trial of gemcitabine plus
Systemic Therapy for Lung Cancer for the Radiation Oncologist 265

cisplatin versus cisplatin alone in patients with locally directed individualized therapy in advanced non-small-cell
advanced or metastatic non-small-cell lung cancer. J Clin lung cancer. J Clin Oncol 25(19):2741–2746
Oncol 18(1):122–130 Smith IE, O’Brien ME, Talbot DC, Nicolson MC, Mansi JL,
Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati Hickish TF et al (2001) Duration of chemotherapy in
A et al (2006) Paclitaxel–carboplatin alone or with advanced non-small-cell lung cancer: a randomized trial of
bevacizumab for non-small-cell lung cancer. N Engl J three versus six courses of mitomycin, vinblastine, and
Med 355(24):2542–2550 cisplatin. J Clin Oncol 19(5):1336–1343
Scagliotti GV (2005) The ALPI Trial: the Italian/European Socinski M, Bondarenko I, Karaseva N, Makhson A,
experience with adjuvant chemotherapy in resectable non- Vynnychenko I, Okamato I et al (2010) Results of a
small lung cancer. Clin Cancer Res 11(13 Pt 2):5011s–5016s randomized, phase III trial of nab-paclitaxel (nab-P) and
Scagliotti G, Monica V, Ceppi P, Righi L, Cambieri A, Volante M carboplatin (C) compared with cremophorbased paclitaxel
et al (2009) Baseline thymidylate synthase expression (P) and carboplatin as first-line therapy in advanced non-
according to histological subtypes of non-small cell lung small cell lung cancer. J Clin Oncol 28(18s)
cancer (NSCLC). J Clin Oncol 27(15s) (Abs# 7521) Socinski MA, Schell MJ, Peterman A, Bakri K, Yates S, Gitten
Scagliotti G, Pastorino U, Vansteenkiste J, Spaggiari L, R et al (2002) Phase III trial comparing a defined duration of
Faccioli F, Orowski T et al (2008a) A phase III randomized therapy versus continuous therapy followed by second-line
study of surgery alone or surgery plus preoperative gem- therapy in advanced-stage IIIB/IV non-small-cell lung
citabine-cisplatin in early stage non-small cell lung cancer: cancer. J Clin Oncol 20(5):1335–1343
Follow-up data of Ch.E.S.T. J Clin Oncol 26(15s):399s Socinski MA, Novello S, Brahmer JR, Rosell R, Sanchez JM,
Scagliotti GV, Parikh P, von Pawel J, Biesma B, Vansteenkiste J, Belani CP et al (2008) Multicenter, phase II trial of sunitinib
Manegold C et al (2008b) Phase III study comparing cisplatin in previously treated, advanced non-small-cell lung cancer.
plus gemcitabine with cisplatin plus pemetrexed in chemo- J Clin Oncol 26(4):650–656
therapy-naive patients with advanced-stage non-small-cell Socinski MA, Langer CJ, Huang JE, Kolb MM, Compton P,
lung cancer. J Clin Oncol 26(21):3543–3551 Wang L et al (2009) Safety of bevacizumab in patients with
Schiller J, Akerley WL, Brugger W, Ferrari D, Garmey E, non-small-cell lung cancer and brain metastases. J Clin
Gerber D et al (2010) Results from ARQ 197-209: A global Oncol 27(31):5255–5261
randomized placebo-controlled phase II clinical trial of Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y,
erlotinib plus ARQ 197 versus erlotinib plus placebo in Ishikawa S et al (2007) Identification of the transforming
previously treated EGFR inhibitor-naive patients with EML4-ALK fusion gene in non-small-cell lung cancer.
locally advanced or metastatic non-small cell lung cancer Nature 448(7153):561–566
(NSCLC). J Clin Oncol 28(18s) Soon YY, Stockler MR, Askie LM, Boyer MJ (2009) Duration
Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, of chemotherapy for advanced non-small-cell lung cancer: a
Krook J et al (2002) Comparison of four chemotherapy systematic review and meta-analysis of randomized trials.
regimens for advanced non-small-cell lung cancer. N Engl J J Clin Oncol 27(20):3277–3283
Med 346(2):92–98 Soria JC, Shepherd FA, Douillard JY, Wolf J, Giaccone G,
Schiller JH, Larson T, Ou SH, Limentani S, Sandler A, Vokes E Crino L et al (2009) Efficacy of everolimus (RAD001) in
et al (2009) Efficacy and safety of axitinib in patients with patients with advanced NSCLC previously treated with
advanced non-small-cell lung cancer: results from a phase II chemotherapy alone or with chemotherapy and EGFR
study. J Clin Oncol 27(23):3836–3841 inhibitors. Ann Oncol 20(10):1674–1681
Scott WJ, Allen MS, Darling G, Meyers B, Decker PA, Putnam Spigel DR, Hainsworth JD, Yardley DA, Raefsky E, Patton J,
JB et al (2010) Video-assisted thoracic surgery versus open Peacock N et al (2009a) Tracheoesophageal fistula forma-
lobectomy for lung cancer: a secondary analysis of data tion in patients with lung cancer treated with chemoradi-
from the American College of Surgeons Oncology Group ation and bevacizumab. J Clin Oncol 27:3385–3390
Z0030 randomized clinical trial. J Thorac Cardiovasc Surg Spigel DR, Greco FA, Burris H, Shipley D, Friedman E,
139(4):976–981 discussion 81–83 Waterhouse D, Whorf R (2009b) A randomized double-
Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, blind placebo-controlled phase II trial of sorafenib and
Costa DB, Heist RS et al (2009) Clinical features and erlotinib or erlotinib in previously treated advanced non-
outcome of patients with non-small-cell lung cancer who small cell lung cancer. J Thorac Oncol 4(9):S355
harbor EML4-ALK. J Clin Oncol 27(26):4247–4253 Stephens RJ, Fairlamb D, Gower NH, Maslove L, Milroy R
Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh (2002) The Big Trial (BLT): Determining the value of
V, Thongprasert S et al (2005) Erlotinib in previously cisplatin-based chemotherapy for all patients with non-
treated non-small-cell lung cancer. N Engl J Med small cell lung cancer. Preliminary results in the supportive
353(2):123–132 care setting. Proc Am Soc Clin Oncol 21(291a)
Siddiqui F, Bae K, Langer CJ, Coyne JC, Gamerman V, Strauss GM, Herndon JE II, Maddaus MA, Johnstone DW,
Komaki R et al (2010) The influence of gender, race, and Johnson EA, Harpole DH et al (2008) Adjuvant paclitaxel
marital status on survival in lung cancer patients: analysis of plus carboplatin compared with observation in stage IB non-
Radiation Therapy Oncology Group trials. J Thorac Oncol small-cell lung cancer: CALGB 9633 with the Cancer and
5(5):631–639 Leukemia Group B, Radiation Therapy Oncology Group,
Simon G, Sharma A, Li X, Hazelton T, Walsh F, Williams C and North Central Cancer Treatment Group Study Groups.
et al (2007) Feasibility and efficacy of molecular analysis- J Clin Oncol 26(31):5043–5051
266 C. P. Belani

Sun SY, Fu H, Khuri FR (2006) Targeting mTOR signaling for Visbal AL, Williams BA, Nichols FC III, Marks RS, Jett JR,
lung cancer therapy. J Thorac Oncol 1(2):109–111 Aubry MC et al (2004) Gender differences in non-small-cell
Tan EH, Salgia R, Besse B, Goss G, Gandara D, Hanna N, lung cancer survival: an analysis of 4, 618 patients
Steinberg J (2009) ABT-869 in non-small cell lung cancer: diagnosed between 1997 and 2002. Ann Thorac Surg
Interim results. J Clin Oncol 27(15S) 78(1):209–215 discussion 15
Thatcher N, Chang A, Parikh P, Rodrigues Pereira J, von Pawel J, Schiller JH, Shepherd FA, Fields SZ, Kleisbauer JP,
Ciuleanu T, von Pawel J et al (2005) Gefitinib plus best Chrysson NG et al (1999) Topotecan versus cyclophospha-
supportive care in previously treated patients with refrac- mide, doxorubicin, and vincristine for the treatment of
tory advanced non-small-cell lung cancer: results from a recurrent small-cell lung cancer. J Clin Oncol 17(2):658–667
randomised, placebo-controlled, multicentre study (Iressa Westeel V, Quoix E, Moro-Sibilot D, Mercier M, Breton JL,
Survival Evaluation in Lung Cancer). Lancet 366(9496): Debieuvre D et al (2005) Randomized study of maintenance
1527–1537 vinorelbine in responders with advanced non-small-cell
Timmerman R, Paulus R, Galvin J, Michalski J, Straube W, lung cancer. J Natl Cancer Inst 97(7):499–506
Bradley J et al (2010) Stereotactic body radiation therapy Winton T, Livingston R, Johnson D, Rigas J, Johnston M, Butts
for inoperable early stage lung cancer. JAMA 303(11): C et al (2005) Vinorelbine plus cisplatin vs. observation in
1070–1076 resected non-small-cell lung cancer. N Engl J Med 352(25):
Vallieres E, Shepherd FA, Crowley J, Van Houtte P, Postmus 2589–2597
PE, Carney D et al (2009) The IASLC Lung Cancer Staging Wozniak AJ, Crowley JJ, Balcerzak SP, Weiss GR, Spiridon-
Project: proposals regarding the relevance of TNM in the idis CH, Baker LH et al (1998) Randomized trial comparing
pathologic staging of small cell lung cancer in the cisplatin with cisplatin plus vinorelbine in the treatment of
forthcoming (seventh) edition of the TNM classification advanced non-small-cell lung cancer: a Southwest Oncol-
for lung cancer. J Thorac Oncol 4(9):1049–1059 ogy Group study. J Clin Oncol 16(7):2459–2465
Combined Radiotherapy
and Chemotherapy: Theoretical
Considerations and Biological Premises
Branislav Jeremic, Dusan Milanovic, and Nenad Filipovic

Contents Abstract
Starting point for full realization of clinical
1 Introduction.............................................................. 267 achievements of combined radiation therapy and
2 Exploitable Mechanisms ......................................... 268 chemotherapy in lung cancer is nothing but full
understanding of general theoretical consideration
3 Implications from Clinical Evidence..................... 269
as well as basic radiobiological premises of
4 Future Consideration .............................................. 271 combined radiation and chemotherapy. Both are
References.......................................................................... 272 nowadays considered as mandatory ingredients in
any consideration of combined radiation therapy
and chemotherapy in lung cancer, as well as
cancers in other tumour sites. More than 30 years
ago, four basic mechanisms through which radia-
tion therapy and chemotherapy can interact were
established including spatial cooperation, indepen-
dent cell kill, protection of normal tissues and
enhancement of tumor response, the latter fre-
quently replaced by terms such as radiosensitiza-
tion or radio enhancement. Clinical evidence
supports importance of exploitable mechanisms,
although protection of normal tissue has never
been proven. Novel radiation therapy technologies,
occasionally changing fractionation pattern and
new drugs/compounds will set the stage and scene
for further verification of basic principles of
combined radiation therapy and chemotherapy in
clinical research of lung cancer.

B. Jeremic (&)
Institute of Lung Diseases, Sremska Kamenica, Serbia
e-mail: nebareje@gmail.com 1 Introduction
D. Milanovic
Department of Radiation Oncology, Over the past several decades combined radiation
University of Freiburg, Freiburg, Germany therapy and chemotherapy have been increasingly
N. Filipovic used in many human cancers. Increasing evidence had
Bioengineering Research Centre, Kragujevac, Serbia been mounting in various tumour sites such as head

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 267
DOI: 10.1007/174_2011_297, Ó Springer-Verlag Berlin Heidelberg 2011
268 B. Jeremic et al.

and neck, cervix, and gastrointestinal cancers, to do not interact negatively to the extent that the overall
name but a few. One of the excellent examples of how level of tumour cell kill is less than that could be
we have progressed as community of oncologists is produced by the best agent, one would expect that two
also a lung cancer case. Here, combination of radia- such agents are more effective than a single agent. In
tion and chemotherapy have also been intensively that scenario, both radiation therapy and chemother-
investigated, particularly in locally advanced stage III apy can act on intrathoracic disease independently
nonsmall cell lung cancer (NSCLC) and limited- leading to cell kill higher than that obtained by the
disease small cell lung cancer. The rationale for better treatment (i.e., radiation therapy) given alone.
combining radiation and chemotherapy in clinic is to This scenario also does not require interaction
achieve improved treatment outcome. This designa- between radiation therapy and chemotherapy.
tion equals ‘‘an improved therapeutic ratio’’, which, in Another mechanism of combined radiation therapy
turn, should be determined as a function both of and chemotherapy in which the combination of the
tumour response and normal tissue damage. Starting two treatments allows delivery of greater dose of
point for full realization of clinical achievements of radiation to be given than would be tolerated other-
combined radiation therapy and chemotherapy in lung wise has been described as Protection of normal tis-
cancer is nothing but full understanding of general sues which could happen only if tumour cells are not
theoretical consideration as well as basic radiobio- similarly protected. In lung cancer, this scenario
logical premises of combined radiation and chemo- would mean that a particular chemotherapy (or any
therapy. Both are nowadays considered as mandatory other) agent protects normal tissue (e.g. oesophageal
ingredients in any consideration of combined radia- mucosa) without protecting an intrathoracic tumour.
tion therapy and chemotherapy in lung cancer, as well This mechanism requests interaction between radia-
as cancers in other tumour sites. tion therapy and chemotherapy. Finally, the mecha-
nism of combination of radiation therapy and
chemotherapy that produces a greater antitumor
2 Exploitable Mechanisms response than would be expected from the response
achieved with the agents used separately has been
More than 30 years ago, four basic mechanisms described as enhancement of tumour response is as
through which radiation therapy and chemotherapy with protection of normal tissues, enhancement of
can interact were established by Steel and Peckham tumour response also requests interaction between
(1979). The situation in which disease in a particular radiation therapy and chemotherapy.
anatomical site that is missed by one therapeutic Any one of these mechanisms by itself could give an
agent is dealt with adequately by another has been improved therapeutic strategy compared with radiation
described as Spatial cooperation. In this scenario, therapy or chemotherapy used alone. Importantly,
radiation therapy would act on disease in thorax (or more than one mechanism may be simultaneously
better said on those parts of the primary tumour and exploited with a particular combination of radiation
the lymph nodes within the radiation therapy treat- therapy and chemotherapy. For example, if one
ment field), but not on extrathoracic (subclinical) attempts to achieve enhancement of tumour response
disease, which should be dealt with by chemotherapy. using chemotherapy agents that have no overlapping
This scenario does not request interaction between the toxicity with radiation therapy, there may also be (1) a
two anticancer agents. The term ‘‘interaction’’ used benefit from the simple addition of anti-tumor effects
here is as it represents the situation in which treatment (independent cell kill), while (2) chemotherapy may
with one agent modifies the response of a tissue also deal with the disease outside the radiation therapy
(normal or tumour) to the second agent. The situation treatment field (spatial cooperation).
which occurs when two partially effective anticancer Of all four possible mechanisms of combined
agents are combined without having to reduce their radiation therapy and chemotherapy, enhancement of
dose levels seriously has been described as Indepen- tumour response was of particular interest for radia-
dent cell kill (simple addition of anti-tumour effects) tion oncologists over the years. In an attempt to
described. With it, an improvement in therapeutic achieve it, different processes may be exploited:
result is sought. Provided that two anticancer agents (a) modification of the initial radiation damage
Combined Radiotherapy and Chemotherapy: Theoretical Considerations and Biological Premises 269

(modification of the slope of the dose–response locally advanced non-small cell lung cancer, such
curves), (b) decreased accumulation or inhibition of studies will not be included into the consideration for
repair of radiation damage in tumour cells, (c) the sake of simplicity of presentation. Examples of
exploitation of induced cell-cycle synchrony (pertur- induction chemotherapy in locally advanced non-
bation in cell kinetics), (d) reoxygenation following small cell lung cancer include studies of Dillman et al.
drug treatment and before irradiation, (e) improved (1990) and Sause et al. (1995) in which induction
drug access following irradiation, (f) decrease of the chemotherapy (two cycles of cisplatin/vinblastine)
tumour bulk by irradiation leading to more rapid was given in a 4 week split followed by radical
proliferation and greater chemosensitivity of tumour radiation therapy (60 Gy in 30 daily fractions) start-
cells, and (g) decrease of the tumour bulk by drugs, ing on day 50, while in the study of Le Chevalier et al.
enabling smaller radiation therapy field-sizes and (1992), a four-drug regimen was given before and
higher radiation therapy doses to be used. after radiation therapy (two cycles each). The ratio-
The enhancement of tumour response may be nale for all the induction chemotherapy studies
additive, infra-additive, or supra-additive (synergis- included decrease of intrathoracic tumour bulk and
tic). Additive effect is the one in which the effect of treatment of subclinical disease outside the thorax.
two independent agents is the sum of the effects that Examples of concurrent radiochemotherapy include a
they would have if acting alone (i.e. 3 ? 2 = 5). study of Schaake-Koning et al. (1992) and studies of
Subadditive effect is the effect of two independent Jeremic et al. (1995), (1996). While the study of
agents which results in a lesser effect than each agent Schaake-Koning et al. (1992) split-course radiation
individually, or the sum of the individual effects (i.e. therapy was used with a total dose of 55 Gy and either
3 ? 2 = 4). The presence of one therapy here weekly (30 mg/m2) or daily (5 mg/m2) cisplatin,
diminishes the effects of the second. Finally, supra- Jeremic et al. used hyperfractionated radiation ther-
additive (synergistic) effect is the effect of two inde- apy with a total radiation dose of either 64.8 Gy
pendent agents which results in a greater effect than (Jeremic et al. 1995) or 69.6 Gy (Jeremic et al. 1996)
each agent individually, or the sum of the individual and concurrent carboplatin/etoposide given either
effects (i.e. 3 ? 2 = 6). The presence of one therapy every other week (Jeremic et al. 1995) or daily
here enhances the effects of the second. (Jeremic et al. 1996). The rationale for the concurrent
A subset of a supra-additive effect would include radiochemotherapy is to improve locoregional (intra-
‘‘sensitization’’ or ‘‘potentiation’’, meaning that of thoracic) tumour control. Although concurrent che-
two agents that are combined one has no effect other motherapy may have acted on the disease outside the
than to increase the effect of the other. Good exam- radiation therapy treatment field, due to its specifics
ples of ‘‘sensitization’’ or ‘‘potentiation’’ include (rather low-doses) it was assumed from the onset that
attempts to overcome tumour hypoxia by using hyp- this type of administration would not have a major
oxic cell sensitizers (e.g. Tirapazamine and RSR-13). effect on systemic disease outside the radiation therapy
treatment field. The aforementioned six studies were
used as examples because they all showed advantages
3 Implications from Clinical Evidence for combined radiation therapy and chemotherapy over
the same radiation therapy given alone. While the
Locally advanced non-small cell lung cancer and induction chemotherapy studies showed a significant
limited-disease small cell lung cancer seem to be survival advantage for the combined approach owing
good examples for verification of theoretical consid- to significant improvement in the distant metastasis
erations in daily clinical practice. The vast majority of control, a finding opposite to that of the concurrent
studies of combined radiotherapy and chemotherapy approach studies, which unequivocally showed sig-
in the two diseases included either induction (upfront, nificant improvement in survival owing to significant
neoadjuvant) chemotherapy followed by radiotherapy improvement in locoregional tumour control.
or concurrent radiation and chemotherapy. Since there If one now wants to put these data into the per-
are only a few studies using alternating radiation spective of exploitable mechanisms of combined
therapy and chemotherapy, or consolidation chemo- radiation therapy and chemotherapy, spatial coopera-
therapy (following concurrent radiochemotherapy) in tion was the only mechanism enabling the therapeutic
270 B. Jeremic et al.

benefit of induction regimens. No independent cell time-scheduled administration of paclitaxel. Their


kill can be noted in any of the induction chemo- results proved to be a possible indicator of radiation
therapy regimens because there was no significant therapy and chemotherapy working together towards
difference in locoregional tumour control, as one enhancement of tumour response. Other two pro-
may have expected if such independent cell kill cesses, which may be of further importance, are
would have happened. Also, no enhancement of improved drug access after radiation therapy as well
tumour response can be noted due to the same rea- as reducing the tumour bulk with radiation therapy
son, due to non-existing overlapping (portions) of leading to more rapid proliferation and greater
the two treatment modalities. Contrary to these chemosensitivity of surviving tumour cells. Although
findings, in concurrent studies, spatial cooperation several prospective phase II studies (Lau et al. 2001;
was not effective, while both independent cell kill Sekine et al. 2006; Gandara et al. 2006) in which
and enhancement of tumour response may have concurrent radiochemotherapy was followed by
occurred. In the low-dose (daily) chemotherapy arms additional (consolidation) chemotherapy showed
of the concurrent studies, however, it seems highly promising results, its first verification in a randomized
unlikely that independent cell kill occurred, due to fashion failed completely. Hanna et al. (2008) showed
its low-doses, which were traditionally considered as no difference in treatment outcome between concur-
less effective than doses usually given with more rent radiochemotherapy alone and the same concur-
split (e.g. every 3 weeks). Therefore, enhancement rent radiochemotherapy followed by consolidation
of tumour response could be seen as the only viable chemotherapy. Their results reconfirmed precious
alternative. observations that concurrent radiochemotherapy
If one now focuses on possible processes exploited without any consolidation chemotherapy (i.e. alone)
in achieving enhancement of tumour response, two of offers the best treatment outcome.
them could have been attractive for induction regi- These clinical examples obtained through pro-
mens, namely (1) shrinking of tumour burden by spective randomized clinical trials show that
drugs enabling smaller radiation therapy treatment exploitable mechanisms of combined radiotherapy
fields and (2) reoxygenation following chemotherapy and chemotherapy in locally advanced NSCLC may
and before radiation therapy. None of these two, be seen as appropriately identified. From the discus-
however, have actually happened in induction che- sion above, one may assume that concurrent radio-
motherapy regimens since there was no significant chemotherapy may offer more possibilities for
difference in locoregional tumour control. In contrast therapeutic benefit, because of involvement of two
to induction chemotherapy regimens, several possible mechanisms, while induction regimens seem to be a
processes seem to have worked well in concurrent good example of only one (spatial cooperation). It is,
radiochemotherapy regimens, such as modification of indeed, what we have recently seen when these two
the initial radiation damage or decreased accumula- treatment approaches have been compared in a pro-
tion or inhibition of repair of radiation damage. Of spective fashion. Albeit of somewhat different study
particular importance is the fact that in all concurrent design, both Japanese study (Furuse et al. 1999, 2000)
radiochemotherapy regimens, the drug was given and a Radiation Therapy Oncology Group study 9410
either before the only daily radiation therapy fraction (Curran et al. 2000) showed an advantage for con-
(Schaake-Koning et al. 1992) or in-between the two current regimens over induction ones due to an
daily fractions (Jeremic et al. 1995, 1996). The aim of improvement in local tumour control (Furuse et al.
these administration modes was to enable drugs to be 2000; Curran et al. 2000).
present in tumour cells at the time of first or second Virtually the same was observed with different
radiation therapy fraction. Therefore, it could both hybrid induction chemotherapy regimens such as
modify the initial radiation damage and decrease/ those using induction chemotherapy followed by
inhibit the repair of radiation damage. Although no concurrent radiochemotherapy with or without con-
prospective randomized trial investigated the effect of solidation chemotherapy. Several clinical trials
induced cell synchrony in NSCLC, a recent study clearly showed that any intensification of the latter/
from Chen et al. (2003) showed excellent results and major part of the combined treatment approach via
promising outcome with radiation therapy and strictly concurrent radiochemotherapy is not effective, once
Combined Radiotherapy and Chemotherapy: Theoretical Considerations and Biological Premises 271

you have started with induction chemotherapy. was 25–26%, and even higher (46%) in very recent
Several studies of Cancer and Leukemia Group B reports of other investigators (Qiao et al. 2005). It is,
(Clamon et al. 1999; Vokes et al. 2002; Akerley et al. therefore, intuitive to identify low-dose daily che-
2005; Socinski et al. 2008) showed that there is no motherapy as the major factor contributing to much
compensation for insufficient start (i.e. with chemo- lower toxicity in contemporary concurrent radioche-
therapy). Other attempts, such as the use of three daily motherapy regimens. Concurrent low-dose chemo-
fractions of radiation therapy (Belani et al. 2005), also therapy in the observation of Jeremic et al.
proved to be ineffective. All in all, whatever you do (unpublished data) also led to grade [3 acute (11%)
after you start with chemotherapy, failure is inevitable and late grade [3 (7%) pulmonary toxicity which
and comes fast. With this approach, you can only compare favourably with those of other series per-
achieve more toxicity (Socinski et al. 2008) and even formed at the same time, and using higher doses of
if you use the modern radiation therapy tools such as concurrent chemotherapy, especially when late bron-
three-dimensional radiation therapy and attempt chopulmonary toxicity is concerned. In the series of
treatment intensification by escalating the total dose, Radiation Therapy Oncology Group, in 170 patients
again, one cannot achieve better outcome. Indeed, treated with hyperfractionated radiation therapy and
impressive 12% mortality in the most recent Cancer concurrent chemotherapy, late grade [3 bronchopul-
and Leukemia Group B attempt (Socinski et al. 2008) monary toxicity was observed in 20% (Byhardt et al.
to combine induction chemotherapy with subsequent 1998).
concurrent radiochemotherapy led investigators to Due to the lack of uniformity in clinical studies
early stopping of the trial. available numerous questions remain, however,
Possible success in combining radiation therapy unanswered. Our reality, unfortunately, includes too
and chemotherapy must be placed into the context of many ‘‘standards’’, too many drugs, and too many
another important aspect, that is, normal tissue mor- questions asked, all inevitably leading to less-than-
bidity (toxicity). Again, sequencing of radiation optimal conclusions and, ultimately, very poor
therapy and chemotherapy (induction vs. concurrent implementation in clinical practice worldwide.
regimens) seems to be a dominating factor. While the
toxicity was usually not troublesome with induction
regimens owing to a gap of a few weeks usually 4 Future Consideration
occurring between the end of chemotherapy and
beginning of radiotherapy, this was one of the major Yet, we have entered the era of strong orientation
concerns in concurrent regimens. What available towards investigation and application of molecular
concurrent studies showed is that the type and mag- biology in this field (Huang and Harari 1999; Brown
nitude of toxicity largely depend on the dose and 2001). The progress and the success in molecular
sequence of chemotherapy administered. Concurrent targeting have formed the basis of many new drug
low-dose chemotherapy led to the incidence of 12% developments. Some of the mainly investigated
acute grade [3 oesophageal toxicity in pooled data compounds include PARP inhibitors, protease inhib-
analysis in patients with stage III NSCLC of Jeremic itors, HDAC inhibitors, m-TOR inhibitors, IGF-IR
et al. (unpublished data) which compares very targeting therapies, COX-2 inhibitors, ALK targeted
favourably to incidence of the same toxicity (and inhibitors, Aurora kinase and Polo-like kinase inhib-
severity) reported during various studies from the itors as well as hedgehog pathway inhibitors. In
same era which used high-dose radiation therapy and addition, agents with a specific targeting moiety
high-dose concurrent chemotherapy. In studies of coupled to a radionuclide (radiopharmaceuticals)
Radiation Therapy Oncology Group (90-15, 91-06, have been investigated. These approaches have pro-
92-04) (Byhardt et al. 1995; Lee et al. 1996; Komaki ven to be effective in ‘‘personalized medicine’’
et al. 1997) when platinum-based doublets were used, regarding lung cancer treatment in recent years.
such incidence ranged 24–53%, being 34% when While majority of the compounds mentioned here
pooled together (Byhardt et al. 1998). In more con- have not yet been investigated in combination
temporary studies, using paclitaxel and carboplatin with radiation within the context of formal phase
(LUN-56 and LUN-63) (Choy et al. 1998, 2000) it I–II studies, several compounds have undergone
272 B. Jeremic et al.

laboratory studies in combination with radiation, and Brown JM (2001) Therapeutic targets in radiotherapy. Int J
have given interesting results while some have even Radiat Oncol Biol Phys 49:319–326
Byhardt RW, Scott CB, Ettinger DS (1995) Concurrent
entered a phase III life (Kishi et al. 2000; Milas 2001; hyperfractionated irradiation and chemotherapy for unre-
Raju et al. 2002). sectable nonsmall cell lung cancer. Results of Radiation
One of the main limitations of current drug ther- Therapy Oncology Group 90-15. Cancer 75:2337–2344
apies, is their high toxicity, which often leads to Byhardt RW, Scott C, Sause WT (1998) Response, toxicity,
failure patterns, and survival in five Radiation Therapy
reduction in the administrable dose, and delivery to Oncology Group (RTOG) trials of sequential and/or
the tumour itself, resulting in a diminished therapeutic concurrent chemotherapy and radiotherapy for locally
effect. One approach considered to circumvent these advanced non-small-cell carcinoma of the lung. Int J Radiat
limitations, is targeted delivery of the therapeutically Oncol Biol Phys 42:469–478
Chen Y, Pandya K, Keng PC, Johnstone D, Li J, Lee Y-J (2003)
active molecule in the effective concentrations to the Phase I/II clinical study of pulsed paclitaxel radiosensitiza-
target organ or tissue where it is needed while tion for thoracic malignancy: a therapeutic approach on the
reducing systemic exposure and unwanted side- basis of preclinical research of human cancer cell lines. Clin
effects. Due to the advantages that nanotechnology Cancer Res 9:969–975
Choy H, Akerley W, Safran H et al (1998) Multiinstitutional
offers, it is expected that nanotechnology will play an phase II trial of paclitaxel, carboplatin and concurrent
important role to cancer treatment in the near future. radiation therapy for locally advanced nonsmall cell lung
However, up to now, the oncologists who used cancer. J Clin Oncol 16:3316–3322
nanoparticles for drug administration usually lacked Choy H, Devore RW 3rd, Hande KR et al (2000) A phase II
study of paclitaxel, carboplatin and hyperfractionated
important information needed to optimally apply this radiation therapy for locally advanced inoperable non-
therapy. It is expected that further research in opti- small-cell lung cancer (a Vanderbilt Cancer Center Affiliate
mizing drug/compound delivery using nanotechnol- Network study). Int J Radiat Oncol Biol Phys 47:931–937
ogy offers substantial advantage over current means Clamon G, Herndon J, Cooper R (1999) Radiosensitization
with carboplatin for patients with unresectable stage III non-
of transportation and drug/compound delivery. small-cell lung cancer: a phase III trial of the Cancer and
If so, this may shed a new light on possible drug- Leukemia group B and the Eastern Cooperative Oncology
related issues (e.g. metabolism, excretion, time to Group. J Clin Oncol 17:4–11
achieve certain plasma level as a surrogate for ther- Curran WJ Jr, Scott C, Langer C, Komaki R, Lee JS, Hauser S
(2000) Phase III comparison of sequential Vs cancer
apeutic efficiency) or time to achieve certain tumor (NSCLC): initial report of Radiation Therapy Oncology
concentration (deemed as necessary for certain level Group concurrent chemoradiation for patients with unre-
of radio enhancement) as well as various radiobio- sected stage III non-small cell lung (RTOG). Proc Am Soc
logical considerations when such new scenario occur Clin Oncol 19:484a (Abstract 1891)
Dillman RO, Seagren SL, Propert KJ, Guerra J, Eaton WL,
with different fractionation regimens, possibly asking Perry MC (1990) A randomized trial of induction chemo-
for new consideration of the four existing exploitable therapy plus high-dose radiation versus radiation alone in
mechanisms of combining radiation therapy and stage III non-small-cell lung cancer. N Engl J Med
chemotherapy in not only lung cancer, but other 323:940–945
Furuse K, Fukuoka M, Kawahara M, Nishikawa H, Takada Y,
cancers as well. Kudoh S (1999) Phase III study of concurrent versus
sequential thoracic radiotherapy in combination with mito-
mycin, vindesine and cisplatin in unresectable stage III non-
References small-cell lung cancer. J Clin Oncol 17:2692–2699
Furuse K, Hosoe S, Masuda N, Sugiura S, Yokota K,
Ohbayashi M (2000) Impact of tumor control on survival
Akerley W, Herndon JE Jr, Lyss AP, Choy H, Turrisi A, in unresectable stage III non-small cell lung cancer
Graziano S, Williams T, Zhang C, Vokes EE, Green MR (NSCLC) treated with concurrent thoracic radiotherapy
(2005) Induction paclitaxel/carboplatin followed by con- (TRT) and chemotherapy (CT). Proc Am Soc Clin Oncol
current chemoradiation therapy for unresectable stage III 19:484a (Abstract 1893)
non-small-cell lung cancer: a limited-access study–CALGB Gandara DR, Chansky K, Albain KS et al (2006) Long-term
9534. Clin Lung Cancer 7:47–53 survival with concurrent chemoradiation therapy followed
Belani CP, Wang W, Johnson DH (2005) Phase III study of the by consolidation docetaxel in stage IIIB non-small-cell lung
Eastern Cooperative Oncology Group (ECOG 2597): cancer: a phase II Southwest Oncology Group study
induction chemotherapy followed by either standard tho- (S9504). Clin Lung Cancer 8:116–121
racic radiotherapy or hyperfractionated accelerated radio- Hanna N, Neubauer M, Yiannoutsos C, McGarry R, Arseneau J,
therapy for patients with unresectable stage IIIA and B Ansari R, Reynolds C, Govindan R, Melnyk A, Fisher W,
non-small-cell lung cancer. J Clin Oncol 23:3760–3767 Richards D, Bruetman D, Anderson T, Chowhan N, Nattam S,
Combined Radiotherapy and Chemotherapy: Theoretical Considerations and Biological Premises 273

Mantravadi P, Johnson C, Breen T, White A, Einhorn L, Qiao WB, Zhao YB, Zhao YH, Wang RZ (2005) Clinical and
Hoosier Oncology Group; US Oncology (2008) Phase III dosimetric factors of radiation induced esophageal injury:
study of cisplatin, etoposide and concurrent chest radiation radiation-induced esophageal toxicity. World J Gastroen-
with or without consolidation docetaxel in patients with terol 11:2626–2629
inoperable stage III non small-cell lung cancer: the Hoosier Raju U, Nakata E, Yang P, Newman RA, Ang KK, Milas L
Oncology Group and U.S. Oncology. J Clin Oncol 26:5755– (2002) In vitro enhancement of tumor cell radiosensitivity
5760 by a selective inhibitor of cyclooxygenase-2 enzyme:
Huang SM, Harari PM (1999) Epidermal growth factor receptor mechanistic considerations. Int J Radiat Oncol Biol Phys
inhibition in cancer therapy: biology, rationale and 54:886–894
preliminary clinical results. Invest New Drugs 17:259–269 Sause WT, Scott C, Taylor S, Johnson D, Livingston R,
Jeremic B, Shibamoto Y, Acimovic L, Djuric L (1995) Komaki R (1995) Radiation Therapy Oncology Group 88–
Randomized trial of hyperfractionated radiation therapy 08 and Eastern Cooperative Oncology Group 4588:
with or without concurrent chemotherapy for stage III non- preliminary results of a phase III trial in regionally
small-cell lung cancer. J Clin Oncol 13:452–458 advanced, unresectable nonsmall cell lung cancer. J Natl
Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1996) Cancer Inst 87:198–205
Hyperfractionated radiation therapy with or without con- Schaake-Koning C, van der Bogaert W, Dalesio O, Festen J,
current low-dose daily carboplatin/etoposide for stage III Hoogenhout J, van Houtte P (1992) Effects of concomitant
non small cell lung cancer: a randomized study. J Clin cisplatin and radiotherapy on inoperable non-small cell lung
Oncol 14:1065–1070 cancer. N Engl J Med 326:524–530
Kishi K, Petersen S, Petersen C, Hunter N, Mason K, Masferrer Sekine I, Nokihara H, Sumi M, Saijo N, Nishiwaki Y, Ishikura
JL (2000) Preferential enhancement of tumor radioresponse S, Mori K, Tsukiyama I, Tamura T (2006) Docetaxel
by a cyclooxygenase-2 inhibitor. Cancer Res 60:1326–1331 consolidation therapy following cisplatin, vinorelbine, and
Komaki R, Scott C, Ettinger D et al (1997) Randomized study concurrent thoracic radiotherapy in patients with unresec-
of chemotherapy/radiation therapy combinations for favor- table stage III non-small cell lung cancer. J Thorac Oncol
able patients with locally advanced inoperable nonsmall cell 1:810–815
lung cancer: Radiation Therapy Oncology Group (RTOG) Socinski MA, Blackstock AW, Bogart JA, Wang X, Munley M,
92–04. Int J Radiat Oncol Biol Phys 38:149–155 Rosenman J, Gu L, Masters GA, Ungaro P, Sleeper A,
Lau D, Leigh B, Gandara D, Edelman M, Morgan R, Israel V Green M, Miller AA, Vokes EE (2008) Randomized phase
(2001) Twice-weekly paclitaxel and weekly carboplatin II trial of induction chemotherapy followed by concurrent
with concurrent thoracic radiation followed by carboplatin/ chemotherapy and dose-escalated thoracic conformal radio-
paclitaxel consolidation for stage III non-small-cell lung therapy (74 Gy) in stage III non-small-cell lung cancer:
cancer: a California Cancer Consortium phase II trial. J Clin CALGB 30105. J Clin Oncol 26:2457–2463
Oncol 19:42–447 Steel GG, Peckham MJ (1979) Exploitable mechanisms in
Le Chevalier T, Arriagada R, Tarayre M, Lacombe-Terrier MJ, combined radiotherapy and chemotherapy: the concept of
Laplanche A, Quoix W (1992) Significant effect of adjuvant additivity. Int J Radiat Oncol Biol Phys 5:85–91
chemotherapy on survival in locally advanced non small Vokes EE, Herndon JE 2nd, Crawford J, Leopold KA, Perry
cell lung carcinoma. J Natl Cancer Inst 84:58 (letter) MC, Miller AA, Green MR (2002) Randomized phase II
Lee JS, Scott C, Komaki R et al (1996) Concurrent chemora- study of cisplatin with gemcitabine or paclitaxel or vino-
diation therapy with oral etoposide and cisplatin for locally relbine as induction chemotherapy followed by concomitant
advanced inoperable non-small-cell lung cancer Radiation chemoradiotherapy for stage IIIB non small-cell lung
Therapy Oncology Group protocol 91–06. J Clin Oncol cancer: Cancer and Leukemia Group B study 9431. J Clin
14:1055–1064 Oncol 20:4191–4198
Milas L (2001) Cyclooxygenase-2 (COX-2) enzyme inhibitors
as potential enhancers of tumor radioresponse. Semin
Radiat Oncol 11:290–299
Radiotherapy and Second Generation Drugs
Michael Geier and Nicolaus Andratschke

Contents Abstract
Unsatisfactory outcomes with radiotherapy alone and
1 Introduction.............................................................. 275 the advent of very potent and possibly radiation
2 Mechanism of Action of Second Generation enhancing drugs, especially platinum compounds
Chemotherapeutic Agents and Their Interaction and vinkaalkaloids, prompted the preclinical and
with Radiotherapy ................................................... 276 clinical investigation of combinded radiochemother-
2.1 Platinum Compounds (Cisplatin and
apy protocols with second generation chemothera-
Carboplatin) ............................................................... 276
2.2 Etoposide.................................................................... 276 peutic drugs as a definitive treatment in inoperable
2.3 Vincaalkaloide (Vinblastin, Vindesine) .................... 277 NSCLC. Not for all substances used unequivocal
2.4 Ifosfamide .................................................................. 277 experimental evidence for a true radiosensitizing
2.5 Mitomycin.................................................................. 277
effect could be demonstrated. Instead, the effects
3 Early Clinical Evaluation ....................................... 277 may result from independent additive cytotoxicity
3.1 Sequential Radiochemotherapy ................................. 278 crucial for sterilizing clonogenic tumor cells sur-
3.2 Concurrent Radiochemotherapy................................ 279
viving radiotherapy and a different non-overlapping
4 Summary................................................................... 287 toxicity profile. This chapter summarizes the pre-
References.......................................................................... 288 clinical data and early clinical evidence for the
rational of combining second generation drugs with
radiotherapy in advanced stage NSCLC.

1 Introduction

Poor outcomes with radiotherapy alone and the advent


of very potent and possibly radiation enhancing drugs,
especially platinum compounds and vincaalkaloids,
prompted an excited interest in the investigation of
combined radiochemotherapy protocols as a definitive
treatment in inoperable NSCLC. Currently, treatment
of choice in inoperable advanced stage non-small-cell
M. Geier  N. Andratschke (&) lung cancer (NSCLC) in curative intent is combined
Department of Radiation Oncology, radiochemotherapy with platinum-based regimens.
Technical University of Munich, Effective protocols have emerged, though outcomes
Munich, Germany still remain unsatisfactory.
e-mail: n_andratschke@yahoo.de

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 275
DOI: 10.1007/174_2011_272, Ó Springer-Verlag Berlin Heidelberg 2011
276 M. Geier and N. Andratschke

Radiotherapy by itself is highly effective in erad- unique metallic element. The oldest in its class,
icating macroscopic tumors and has a higher level of Cisplatin, still represents the most widely used and
efficacy in quantitative tumor cell kill compared to effective drug in chemoradiation. Its potent antitumor
chemotherapy alone. Macroscopic tumors especially effects are based on the inhibition of DNA synthesis
at an early stage without lymph node involvement and the formation of DNA interstrand cross-links. It
(cT1-3 cN0 cM0) can be controlled by radiotherapy has long been recognized as a potent radio-sensitizing
alone. While radiotherapy has the efficacy to destroy agent both in vitro and in vivo, though supra-additive
all clonogenic cells of a macroscopic solid tumor, effects have not been consistently demonstrated in all
chemotherapy can only reduce the total number of experimental studies. The mechanism of interaction
tumor cells by 2–3 orders of magnitude. Though this has not fully been elucidated. It has been assumed to
tumor cell kill is not sufficient for permanent local inhibit PLDR and SLDR (Douple and Richmond
tumor control by itself, it may be crucial in combined 1978) as well as act preferentially on hypoxic tumor
modality treatment with radiation therapy improving cells rendering them more susceptible to radiation
overall response and eventually survival by sterilizing (Ziegler and Kopp 1987). Still, the radiation
clonogenic tumor cells surviving radiotherapy. In enhancing properties may well mainly rely on its
addition, chemotherapy may render clonogenic tumor independent cytotoxicity adding to the effect of the
cells more susceptible to radiation therapy increasing combined treatment and the different toxicity profile
the net cell kill compared to radiotherapy alone by not overlapping with radiation-induced toxicity.
modulating processes involved in tumor radioresis- Regardless of the mechanism involved, it has been
tance such as sub-lethal damage repair, repopulation shown that a close temporal interaction is necessary to
or intrinsic radiosensitivity. Furthermore chemother- maximize the combined effects of radiotherapy and
apy has the potential to control an early stage of Cisplatin. Optimal results with regard to radiation
systemic cancer disease by sterilizing microscopic enhancement have been observed when administered
metastases. This is an important aspect in the treat- with fractionated radiotherapy shortly before each
ment of advanced stage NSCLC by chemoradiation. fraction (Dewit 1987). Carboplatin is a newer plati-
Therefore chemotherapy has become an integral part num derivative specifically lacking the renal toxicity
in combined cancer treatment, especially in the seen with Cisplatin. Exerting similar radiation
treatment of advanced stage NSCLC. An associated enhancing effects with a favorable toxicity profile
higher acute toxicity caused by interaction of these readily allowed for an effective introduction into
two treatments has to be considered, particularly for clinical chemoradiation protocols.
concurrent chemoradiotherapy. Especially the risk of
late treatment toxicity should be kept at an acceptable
limit. 2.2 Etoposide
This chapter summarizes the pre-clinical data and
early-clinical evidence for the rational of combining Topoisomerases are a class of enzymes necessary to
second generation drugs with radiotherapy in maintain the integrity of the genome during tran-
advanced stage NSCLC. scription, replication and recombination by acting on
DNA topology (Binaschi et al. 1995). Etoposide as an
inhibitor of topoismerase II leads to stable and thus
2 Mechanism of Action of Second irreparable DNA single- and double-strand breaks
Generation Chemotherapeutic (ratio approx. 10–20:1) by stabilizing the DNA
Agents and Their Interaction cleavage complex preventing religation of DNA.
with Radiotherapy Radiation-induced single-strand breaks are converted
to double-strand breaks in the presence of toposi-
2.1 Platinum Compounds (Cisplatin merase inhibitors, especially in S phase of the cell
and Carboplatin) cycle (Chen et al. 1997). This way, Etoposide leads
to a significant increase in lethal double-strand
Platinum-based compounds form a distinct class of breaks and hence to an increased cytotoxicity of the
very potent anti-cancer agents characterized by its combined treatment.
Radiotherapy and Second Generation Drugs 277

2.3 Vincaalkaloide (Vinblastin, clonogenic survival. At the S-phase of the cell cycle it
Vindesine) has been suggested that especially the enhancement of
radiation effects could be mediated due to interference
Vincaalkaloids—originally derived from Catharan- of Ifosfamide with the repair pathways of radiation-
thus roseus/vinca rose—are a class of drugs that induced potentially lethal damage (Latz et al. 1998;
interfere with the formation and function of micro- Latz and Weber 2002).
tubuli by interacting with tubulin, a microtubular
protein of the mitotic spindle apparatus necessary for
cell division. As microtubuli destabilizing agents bind 2.5 Mitomycin
soluble and microtubuli-associated tubulin and thus
inhibit a correct configuration of the mitotic spindle, Mitomycin, a bioreductive alkylating agent isolated
they act particularly on proliferating cells. from Streptomyces caespitosus or Streptomyces lav-
At low concentrations cell cycle arrest in meta- endulae, has been initially used as an antibiotic before
phase is induced. At higher concentrations microtu- its antitumor activity was recognized. Reductive
bule depolymerization and mitotic spindle destruction activation of the drug is required to bind DNA by
are observed. The antitumor activity of Vinblastine mono- or bifunctional alkylation which causes
and Vindesine is thought to be primarily due to these cross-linking of the DNA strands with high efficiency
cell cycle phase-specific interactions (Jordan et al. and leads to inhibition of DNA synthesis and function
2004). Subsequently cell death occurs by several (Verweij and Pinedo 1990).
mechanisms from different forms of mitotic slippage Due to this mechanism Mitomycin C preferentially
or adaptation to mitotic catastrophe. Vindesine, a kills hypoxic tumor cells. Assuming that normal tissues
semi-synthetic vinca alkaloid, derived of Vinblastine, are less hypoxic, the selective targeting of this cell
is three times more potent than Vincristine and nearly population has the potential to improve tumor cure
10 times more potent than Vinblastine in causing without compromising normal tissue complication
mitotic arrest in vitro. rates, though this differential effect seems only relevant
The hypothetical benefit of combining radiother- for a very low oxygen tension.
apy with the Vinblastine or Vindesine, i.e. supra- In vivo a significant enhancement of radiation
additive cell kill due to the cell cycle arrest at the response of solid tumors could be shown in pre-
radiosensitive M-phase, could not conclusively be clinical studies administering Mitomycin C before
demonstrated in pre-clinical studies (Sui et al. 2005; radiation. The enhancement seemed to be related both
Van Belle et al. 1994). to direct radiosensitization and independent cytotox-
icity against radio-resistant hypoxic cells (Grau and
Overgaard 1991).
2.4 Ifosfamide

The small molecule Ifosfamide belongs to the group 3 Early Clinical Evaluation
of alkylating agents. The exact mode of action by now
is not entirely clear, but it is most likely similar to the Chemotherapy can be administered before, during or
other agents of that group. After biotransformation in after radiotherapy. The aims of the concepts are very
the liver by the cytochrome P450 oxygenases the different. Sequential chemoradiotherapy adding
active metabolites of the pro-drug alkylate or bind radiotherapy after chemotherapy treatment is used to
with many intracellular molecular structures, includ- increase the rate of clonogenic cell kill in critical
ing nucleic acids. The main cytotoxic action is caused areas with micro- or macroscopic tumor residuals that
by the alkylation of DNA with formation of inter and are not treated sufficiently by the preceding chemo-
intra strand cross-links in the DNA that lead to cell therapy. Concurrent chemoradiation may have several
death. (Fleming 1997). advantages compared to the sequential protocols
In vitro supra-additive effects could be shown besides independent cytotoxicity as it combines the
after exposure of different human cancer cell lines effects of spatial and temporal cooperation to increase
to Ifosfamide combined with radiation assessed by net tumor cell kill via additive or even supra-additive
278 M. Geier and N. Andratschke

effects and avoids the detrimental effect of treatment of chemotherapy in this manner resulted only in an
prolongation and possible induction of repopulation increase of acute toxicity with no beneficial effects to
by chemotherapy. response rates/progression-free and overall survival
The clinical phase I/II studies presented in the with an observed median survival of 12 months in both
following sections reflect quite nicely the aforemen- arms and median time to progression of 30 weeks for
tioned different rationales for combining second chemoradiation and 35 weeks for radiotherapy alone
generation drugs with radiotherapy either in a (Planting et al. 1996).
sequential or a concurrent setting with or without In the GOTHA I trial hyperfractionated accelerated
induction or consolidation chemotherapy. Particularly radiotherapy was combined with simultaneous and
the concurrent radiochemotherapy protocols employ- adjuvant chemotherapy alternating Cisplatin and
ing low-dose platinum-based regimens were designed Vinblastin. Radiotherapy was given in 1.5 Gy single
to exploit synergistic effects by close temporal dose in 42 fractions with b.i.d. treatment during weeks
cooperation as a results of daily or weekly adminis- 2, 3, 6 and 7 to a total dose of 63 Gy. Chemotherapy
tration at presumed sub-toxic dose levels shortly consisted of six cycles Cisplatin 70 mg/m2 (day 1)
before each fraction while aiming at reducing acute and Vinblastin 5 mg/m2 (day 7) during week 1 and 5
toxicity, especially acute esophageal reaction. overlapping with radiotherapy, and week 9, 13, 17,
and 21 afterwards. The long term results of the Suisse
study including 67 inoperable stage III patients were
3.1 Sequential Radiochemotherapy published together with those of the consecutive
GOTHA II trial that evaluated the same hyperfrac-
An Italian phase II trial evaluated sequential chemo- tionated accelerated radiotherapy combined with a
radiation with three cycles of high-dose chemotherapy different alternating chemotherapy including three
consisting of Ifosfamide 3 g/m2 day 1, Carboplatin cycles of Cisplatin 60 mg/m2 and Mitomycin 8 mg/
200 mg/m2 days 1 and 2, Etoposide 120 mg/m2 days m2 on day 1 and Vindesine 3 mg/m2 on days 1 ? 8
1–3 as well as rhG-CSF support followed by normo- during weeks 1, 5, 9, 13, 17 and 21. The analysis with
fractionated radiotherapy to a total dose of 60 Gy. a minimum follow-up of 3 years and an median
Only 61% of the 61 enrolled patients completed the follow- up for survivors of 6 years showed a 1-, 3-,
intended chemoradiation regimen. The results regard- 5- and 8-year overall survival of 56, 27, 12 and 9%
ing survival were disappointing with a median with a median survival of 13.6 months. No significant
progression-free survival of 5.4 months and a median differences in survival for stage IIIA versus IIIB or
1-year overall survival rate of 31%. Toxicity was performance status or the two treatment arms could
modest and mainly due to grade 3 or greater thrombo- be detected. Long time survival does not strongly
cytopenia in 24% of the administered chemotherapy correlate with response rates. Acute toxicity consisted
cycles (Scagliotti et al. 1996). mainly of grade 3 or greater hematological side
A phase II EORTC study compared response rates effects with lower rates of non-hematological toxicity
and morbidity of sequentially combined high dose split (e.g. mucositis and nausea) and very low rates of
course radiotherapy with induction chemotherapy to grade 3–4 lung toxicity. Four treatment-related deaths
high dose split course radiotherapy alone. Induction were reported (Mirimanoff et al. 1998).
chemotherapy consisted of Cisplatin 100 mg/m2 on The GOTHA II trial included 65 Patients treated
days 1 and 22 and Vindesine 3 mg on days 1 and 8, and with split course hyperfractionated accelerated
22 and 29. Hypofractionated split-course radiotherapy radiotherapy to a total dose of 63 Gy in 1.5 Gy b.i.d.
of 30 Gy in 3 Gy per fraction and 25 Gy per 2.5 Gy in five fractions a week combined with an alternating
fraction separated by a 21 days interval to a total dose chemotherapy including three cycles of Cisplatin
of 55 Gy was administered beginning with day 1 in the 60 mg/m2 and Mitomycin 8 mg/m2 on day 1 and
radiotherapy arm and day 43 in the chemoradiation arm Vindesine 3 mg/m2 on days 1 ? 8 during weeks 1, 5,
with a third cycle of chemotherapy between the two 9, 13, 17 and 21. Superior results regarding survival
radiotherapy courses. Of 70 randomized patients with with an acceptable increase of toxicity compared to
stage III NSCLC 34 were evaluated in the chemoradi- conventional radiotherapy alone were reported by the
ation arm and 30 in the radiotherapy arm. The addition suisse group (Mirimanoff et al. 1998).
Radiotherapy and Second Generation Drugs 279

The Japanese Phase II study JCOG 9306 assessed In an earlier phase II Southwest Oncology Group
the efficacy and toxicity of alternating chemoradiation protocol standard fractionated thoracic irradiation to a
combining hyperfractionated accelerated radiotherapy total dose of 61 Gy combined with concurrent daily
to a total dose of 66–70 Gy with 1.5 Gy b.i.d. with low-dose Cisplatin (5 mg/m2) followed by three
Cisplatin 80 mg/m2 on day 1 and Vindesine 3 mg/m2 cycles of high-dose Cisplatin consolidation chemo-
on days 1 and 8. Radiotherapy was given during therapy (100 mg/m2 on day 1 and 8) was studied for
weeks 1, 2, 5, 6 and 9, whereas chemotherapy during feasibility and effectiveness in 64 eligible patients
week 3 and 7 and optionally week 10. Of 41 enrolled with locally advanced unresectable NSCLC. 1- and
patients 32 completed the intended treatment. In 7 2-year actuarial survival rates of 56 and 24% and a
of the 41 Patients chemotherapy was discontinued median survival for all patients of 14 months were
before completing at least two cycles, due to reported. Stage IIIa patients showed a better median
treatment-related toxicity (3), patients refusal (1), survival of 17 months and a 2 year survival rate of
progressive disease (1) or complications unrelated to 38%, as compared with 10 months and 14% for stage
treatment. Radiotherapy could not be completed IIIb patients, respectively. 8% of the Patients could
because of treatment-related toxicity, progressive not complete concurrent chemoradiation due to
disease and complications in 3, 1 and 2 patients, toxicity. Acute grade 3 side effects were reported for
respectively. Median 3- and 5-year survival rates of esophagitis (16%), leukopenia (14%), nausea (8%),
24 and 10% and a median survival of 18.4 months and vomiting (6%) (Hazuka et al. 1994).
were accompanied by at least grade 3 leucopenia and In the EORTC 08912 phase I/II study Uitterhoeve
esophagitis in 32 and 7 patients, respectively and late et al. investigated the feasibility of radiotherapy and
esophageal toxicity of at least grade 3 in 2 patients. chemotherapy dose escalation given as concurrent
In most of the patients therapy had to be paused chemoradiation in 40 inoperable NSCLC patients.
due to the resulting acute hematological side effects Dose escalation was carried out at four different levels
(Sekine et al. 2002). increasing the total doses of a simultaneous integrated
boost radiotherapy subsequently from 55 Gy in 2.75
per fraction to 60.5 Gy giving two extra fractions at
3.2 Concurrent Radiochemotherapy levels I and II and to 66 Gy at levels III and IV giving
four extra fractions, respectively. Furthermore dose
3.2.1 Single Agent Regimens escalation of concurrent daily Cisplatin (6 mg/m2)
was carried out beginning at level II and III com-
3.2.1.1 Cisplatin bining two extra fractions of radiotherapy and four
The RTOG trial 92-04 compared induction chemo- extra fractions at Level IV respectively with concur-
therapy with Vinblastine and Cisplatin followed by a rent Cisplatin. The authors reported good overall
concurrent chemoradiation with lower dose Cisplatin survival rates at 1 and 2 years with 53 and 40%,
every 2 weeks simultaneous with hyperfractionated respectively. This survival was associated with rather
accelerated radiotherapy (HART) with concurrent low side effects regarding C grade 3 acute toxicity with
chemoradiation consisting of Cisplatin and Etoposide nausea in 8% and leucopenia and thrombocytopenia in
and the same dosed HART (69.6 Gy 1.2 Gy per 5% of the patients. Late toxicity for esophageal
fraction b.i.d.). Overall survival was the primary symptoms C grade 3 was reported in 5% with only
endpoint of the study. For the 80 included patients grade 1 and 2 radiation pneumonitis in 3% of the
treated with the Cisplatin mono scheme the 1-year patients, respectively (Uitterhoeve et al. 2000).
overall survival was 65% with a median survival of An triple agent induction chemotherapy protocol
15.5 months and an 1-year progression-free survival with Ifosfamide, Etoposide and Cisplatin preceding
rate of 50%. Predominantly hematological grade 4 thoracic radiotherapy combined with continuous
toxicity was reported in that arm similar to the earlier low-dose Cisplatin infusion (6 mg/m2/days) for
RTOG trial. Non-hematological acute and late side stage IIIb NSCLC patients was evaluated by Pujol
effects consisted mainly of esophageal toxicity with et al.. Induction chemotherapy consisted of three
grade III or higher acute toxicity in 6% and late courses of Ifosfamide 1.5 g/m2, Etoposide 100 mg/m2
toxicity in 3% of the patients (Komaki et al. 1997). and Cisplatin 25 mg/m2, given on days 1–4 of a
280 M. Geier and N. Andratschke

21 day cycle. Recombinant human methionyl granu- chemotherapy consisting of Cisplatin, Mitomycin and
locyte colony stimulating factor was used for hema- a vinca alkaloid was followed by a standard frac-
topoietic support. A split-course normo-fractionated tionated thoracic radiotherapy to a total dose of
thoracic radiotherapy (first course: 30 Gy/10; 4 week 62–66 Gy with concurrent daily bolus or continuous
rest period; second course: 25 Gy/10) and concurrent infusion of Carboplatin. A median survival of 12
continuous low-dose Cisplatin infusion of 6 mg/m2 months was reported for the 29 included patients. As
daily was administered to patients with at least stable the dose escalation of Carboplatin showed limiting
disease after induction chemotherapy. This rather thrombocytopenia for 15 mg/m2/days, 10 mg/m2
aggressive protocol with intense induction chemother- were recommended by the authors when used in this
apy with high therapy-related toxicity and split-course setting (Thomas et al. 1997).
radiotherapy did yield a disappointing outcome regard- Dose escalation of Carboplatin concurrent to
ing locoregional progression and overall survival (Pujol accelerated hyperfractionated radiotherapy to a total
et al. 1999). dose of 60 Gy in 1.5 per fraction b.i.d. followed by
Another low-dose Cisplatin-based chemoradio- four cycles of consolidating Carboplatin chemother-
therapy regime using a triple induction chemotherapy apy (350 mg/m2) was assessed in an phase I study of
protocol with Vindesine, Ifosfamide and Cisplatin and Kelly et al. including 30 patients with inoperable
subsequent Cisplatin concurrent to radiotherapy was NSCLC. Chemotherapy consisted of daily Carbo-
assessed by a Belgium phase II trial. From 1993 to platin 25 mg/m2 or 30 mg/m2 concurrent to radio-
1995 23 stage III NSCLC patients were enrolled to therapy. Two of six patients receiving 30 mg/m2
the study. Three cycles of Cisplatin 30 mg/m2 and experienced grade 4 dose-limiting esophagitis, in
Ifosfamide 1,200 mg/m2 on days 1–3 plus Vindesine contrast to 3 of 24 patients in the 25 mg/m2 group.
3 mg/m2 on days 1 and 8 were administered every During consolidation chemotherapy grade 4 throm-
4 weeks followed by an hypofractionated radiother- bocytopenia was observed in one of 22 patients.
apy to a dose of 30 Gy in 3 Gy per fraction and a Median 1- and 2-year survival rates of 63 and 49%,
boost to an accumulative total dose of 52.2 Gy respectively, were reported by the author with an
in 2.2 Gy with every fraction preceded by daily median survival of 18.3 months (Kelly et al. 1998).
Cisplatin 6 mg/m2. In four patients induction could The feasibility and efficacy of concomitant
not be completed due to one therapy-related death and radiochemotherapy with standard fractionated radio-
progressive disease in three patients and subsequent therapy to a total dose of 66 Gy and concurrent daily
radiotherapy was not administered. This Ifosfamide Carboplatin chemotherapy with 15 mg/m2 adminis-
containing chemoradiotherapy protocol showed dis- tered after two cycles of induction chemotherapy
appointing survival. Treatment was accompanied by (on day 1 and 28) with Etoposide (100 mg/m2) on
quite high toxicity which was predominantly of days 1 to 3, and Carboplatin (350 mg/m2) on day
hematologic origin, but also neurologic and pulmonal 1 was evaluated in a phase II trial by Bardet et al.
grade 3 or greater toxicity was observed (Van den Fourty patients with locally advanced unresectable
Brande et al. 1998). non-metastatic NSCLC with at least stable disease
Several other smaller phase I/II studies have also after induction chemotherapy were treated with
investigated the feasibility and effectiveness of daily above described concurrent chemoradiotherapy
low-dose Cisplatin as a single agent concurrent to starting on day 55 followed by two additional cycles
radiochemotherapy. As the study design and the of Etopopside and Carboplatin for 4 weeks after
results are similar to the above-mentioned studies, completion of chemoradiotherapy. Thirty eight per-
they have been included in Table 1 for the sake of cent of the 37 patients who received full-induction
completeness. chemotherapy showed grade 3–4 hematological
toxicity with a response rate of only 11%. Twelve of
3.2.1.2 Carboplatin 26 non-progressive patients received the additional
Thomas et al. reported on a phase I study eval- two cycles of Etoposide and Carboplatin. Median
uating Carboplatin as radiosensitizer after Cisplatin- survival was only 9 Months with 1 and 2 year
based induction chemotherapy in NSCLC patients overall survival rates of 38 and 15%, respectively
with locally advanced disease. Induction of triple (Bardet et al. 1997).
Table 1 Summary of phase I/II studies with second generation chemotherapy concurrent to radiotherapy
Author Number RTx CTx Overall 1 year 2 years 5 years Hematological Non- Remarks
of survival, (%) (%) (%) toxicity C3 hematological
patients median (%) toxicity C3
(months) (%)
Komaki et al. 80 Standard Cisplatin 15.5 65 62 (grade 4) Esophageal 3 Induction:
(1997) fractionated Cisplatin,
Vinblastine
Van den 19 Hypofractionated Cisplatin l.d. 10.6 47 16 Leukopenia 22 Neurotoxicity Induction:
Brande et al. daily 18; Cisplatin,
(1998) pneumonitis Ifosfamide,
11 Vindesine
Pujol et al. 44 Hypofractionated Cisplatin l.d. 12 49 19 5 Neutropenia 66 Late Induction:
(1999) split-course daily (3 years) (grade 4), pulmonary 23, Cisplatin,
Radiotherapy and Second Generation Drugs

leukopenia 34 esopahgeal 9 Ifosfamide,


(grade 4), Etoposide
thrombopenia
25 (grade 4)
van 40 Hypofractionated Cisplatin l.d. 10.5 Thrombopenia –
Harskamp split-course daily 2
et al. (1987)
Boven et al. 40 Hypofractionated Cisplatin l.d. 10.5 – –
(1988) split-course daily
Hazuka et al. 65 Standard Cisplatin l.d. 14 56 24 14 Esophageal Consolidation:
(1994) fractionated daily 16, nausea 8 Cispaltin;
(grade 3),
nausea 6
(grade 4)
Palazzi et al. 36 Hyperfractionated Cisplatin l.d. 8 37 – Esophageal Continuous
(1996) accelerated daily 14, late concomittant:
pulmonary 3 Cisplatin
(grade 5)
Sarihan et al. 15 Concomittant Cisplatin l.d. 16 – Nausea 60
(1998) boost daily
Ardizzoni 32 Standard Cisplatin l.d. 12.5 52 26 19 21 (RCTx), 31 Esophageal 6 Induction:
et al. (1999) fractionated daily (3 years) (Induction Cisplatin,
CTx) Vinblastine
(continued)
281
Table 1 (continued)
282

Author Number RTx CTx Overall 1 year 2 years 5 years Hematological Non- Remarks
of survival, (%) (%) (%) toxicity C3 hematological
patients median (%) toxicity C3
(months) (%)
Uitterhoeve 40 Hypofractionated Cisplatin l.d. 53 40 10 Nausea/ Phase I/II;
et al. (2000) daily vomiting 8,
late
esophageal 5
Thomas et al. 29 Standard Carboplatin 12 – Esophageal 10 Phase I
(1997) fractionated l.d. daily (at highest induction:
Carboplatin Cisplatin,
level of dose Mitomycin,
escalation) Vinca Alkaloid
Bardet et al. 40 Standard Carboplatin 9 38 15 38 (induction 21 Induction:
(1997) fractionated l.d. daily CTx), 13 Carbopaltin,
(RCTx) Etoposide;
Kelly et al. 30 Hyperfractionated Carboplatin 18.3 63 49 4 Esophageal 43 Consolidation:
(1998) accelerated l.d. daily (consolidation (grade 4) Carboplatin
CTx)
Kalemkerian 19 Standard Cisplatin l.d. 18 42 11 47 (RCTx), 38 Esophageal 42 Phase I;
et al. (1999) fractionated daily, (consolidation consolidation:
Etoposide CTx) Cisplatin,
l.d. daily Etoposide
Lee et al. 76 Hyperfractionated Cisplatin, 18.9 67 35 57 (grade 4) 6.6 (grade 4/5),
(1996, 1998) accelerated Etoposide esophageal 53,
pulmonary 25
Komaki et al. 82 Hyperfractionated Cisplatin, 14.4 58 33 (grade 4) Esophageal 38
(2002) accelerated Etoposide
Pottgen et al. 30 Hyperfractionated Cisplatin, 13 31 Leukopenia 63, Esophageal 33, Induction:
(2002) accelerated, Etoposide Thrombopenia Pneumonitis 3 Cisplatin,
standard 23 (grade 4) Etoposide
fractionated boost
Blanke et al. 21 Standard Cisplatin, 50.2 (weeks) 45 Leukopenia 55, Pulmonary 25, Early closure
(1997) fractionated Etoposide thrombopenia esophageal 40 due to
40 (grade 4) pulmonary
toxicity
(continued)
M. Geier and N. Andratschke
Table 1 (continued)
Author Number RTx CTx Overall 1 year 2 years 5 years Hematological Non- Remarks
of survival, (%) (%) (%) toxicity C3 hematological
patients median (%) toxicity C3
(months) (%)
Albain et al. 50 Standard Cisplatin, 15 17(3 year) 15 Neutropenia 32 Esophageal 12 Consolidation:
(2002) fractionated Etoposide (grade 4) (grade 3), Cisplatin,
esophageal 8 Etoposide
(grade 3)
Lau et al. 63 Standard Carboplatin, 13 21 Leukopenia 50, Esophageal 15 Poor risk
(1998) fractionated Etoposide tthrombopenia patients
23
Jeremic et al. 58 Hyperfractionated Carboplatin, 10 42 24 9.1 22 Esophageal 7, Elderly
(1999) accelerated Etoposide Pulmonary 4 patients
Radiotherapy and Second Generation Drugs

(oral)
Jeremic et al. 41 Hyperfractionated Carboplatin, 25 34(3 years) 29 30 Esophageal
(1999) accelerated Etoposide 15, Pulmonary
12
Tsuchyia 11 Hyperfractionated Cisplatin, 64 Leukopenia 91 Esophageal 27 Phase I
et al. (2001) accelerated Vindesine
10 Standard Cisplatin, 70 Leukopenia 90 Pneumonitis
fractionated Vindesine 20 (grade5)
Kubota et al. 70 Standard Cisplatin, 14.8 14.8 Leukopenia 93 Nausea/
(2000) fractionated Vindesine vomiting 27
split-course
Le Pechoux 34 Hyperfractionated Low-dose 53 12 6 (grade 4) Esophageal 9 Consolidation:
et al. (1996) accelerated Cisplatin, Cisplatin,
Vindesine Vindesine;
Byhardt et al. 42 Hyperfractionated Cisplatin, 12.2 54 28 45 (grade 4) Esophageal 24
(1995) accelerated Vinblastine
Furuse et al. 61 Standard Cisplatin, 16 36.7 Leukopenia 95, Nausea 16,
(1995) fractionated Mitomycin, Thrombopenia Esophageal 6
split-course Vindesine 45
Atagi et al. 22 Standard Cisplatin, 19 84.8 34.5 Leukopenia 82, Esophageal 5,
(2002) fractionated Mitomycin, thrombopenia pulmonary 5
Vindesine 27 (grade 4)
Lee et al. 161 Hyperfractionated Cisplatin, 15 51.2 25.1 14.8 – –
(2003) accelerated Mitomycin,
Vinblastine
283

l.d. low-dose
284 M. Geier and N. Andratschke

3.2.2 Double Agent Regimens 2 years; 30 versus 49% at 4 years). Similar survival
rates with 5-year overall survival rates of 16% and a
3.2.2.1 Cisplatin/Etoposid median survival of 15.5 months compared to 13% and
There are several phase I and II studies evaluating 16.4 months in the standard arm of the study were
concurrent chemoradiation combining Cisplatinum observed. Non-hematological side effects were pre-
and the topoisomerase-II-inhibitor Etoposide with dominantly observed in the hyperfractionated arm,
either standard fractionated or hyperfractionated especially with regard to acute and late esophageal
accelerated radiotherapy that showed promising toxicity, though dose-reduced Etoposide led to a
results compared to radiotherapy alone or even other decreased acute esophageal toxicity compared to the
platinum-based chemotherapy regimens. RTOG 91-06 trial, C grade 3 toxicity 38 versus 53%,
In the phase II RTOG trial 91-06 two cycles of respectively (Komaki et al. 2002).
Etoposide (50 mg day 1 alternating with 50 mg b.i.d. The SWOG 9019 phase II evaluated chemora-
days 1–21) and Cisplatin (40 mg/m2 on day 1 and 8 diotherapy with Cisplatin (50 mg/m2) on days 1, 8,
of a 28 day cycle) were administered concurrent 29, 33 and etopsoide (50 mg/m2) on days 1–5,
to hyperfractionated accelerated radiotherapy with 29–33 concurrent to standard fractionated radio-
1.2 Gy b.i.d to a total dose of 69.6 Gy. Estimated 1- therapy to a dose of 45 Gy in 1.8 Gy per fraction,
and 2-year overall survival was 67 and 35%, respec- followed by a boost to a total dose of cumulative
tively and with a median survival of 18.9 months 61 Gy in 2 Gy per fraction and additional two
quite promising. A subgroup of patients with less than cycles of Cisplatin and Etoposide as consolidation
5% weight loss had even better survival rates with an chemotherapy for 50 patients with pathologically
estimated 1- and 2-year survival of 70 and 42% and a proven stage IIIB NSCLC. Boost treatment and
median survival of 21.1 month. Acute toxicity was consolidation chemotherapy was administered
considerable with grade 4 hematologic side effects in exclusive to patients with at least stable disease and
57% of patients. Non-hematological acute toxicities no change in pulmonary function. The study
higher than grade 3 were observed in 53 and 25% revealed an overall median survival of 15 months
regarding symptoms of the esophagus and lung, with 3 and 5 year survival rates of 17 and 15%,
respectively. These early phase II results could be respectively. The most frequent therapy-related
confirmed by a later published report on long-term toxicity was hematological with grade 4 neutropenia
follow-up. After 5 years of minimum follow-up 5 of in 32% of the patients. Major non-hematological
18 eligible patients were still alive with no evidence toxicity was observed for esophagitis with grade
of disease. A 5 year survival rate of 22% was repor- 3 and 4 symptoms in 12 and 8% of the patients,
ted. One treatment-related death due to severe radia- respectively (Albain et al. 2002).
tion pneumonitis was reported (Lee et al. 1996, 1998). In a more recent German phase I/II trial, including
Due to the high treatment-related toxicity observed 30 stage III NSCLC patients two cycles of induction
in the RTOG 91-06 trial, especially esophageal chemotherapy consisting of Cisplatin (60 mg/m2/
toxicity, a subsequent randomized phase II RTOG day on days 1 and 7) and Etoposide (150 mg/m2/
trial 92-04 tested the same treatment with reduced day on days 3–5) followed by chemoradiation with
doses of Etoposide (50 mg b.i.d on days 1–10) and one cycle of Cisplatinum (50 mg/m2/day on days 1
Cisplatin (50 mg/m2 on day 1 and 8) concurrent to and 7) and Etopside (100 mg/m2/day on days 3–5)
hyperfractionated accelerated radiotherapy. This was evaluated with regard to overall survival and
arm was compared to a treatment regime of induc- local control. Radiation treatment was given as
tion chemotherapy with Vinblastine and Cisplatin hyperfractionated accelerated radiotherapy to a dose
followed by a chemoradiation with lower doses of of 45 Gy in 1.5 Gy b.i.d. followed by a normo-
Cisplatin every 2 weeks of daily radiotherapy to a fractionated boost of 20 Gy to a total cumulative dose
total dose of 63 Gy in 1.8 Gy per fraction. Treatment of 65 Gy. Acceptable toxicity with no treatment-rela-
arm with concurrent chemotherapy and hyperfrac- ted deaths was reported with grade 3 and 4 esophagitis
tionated accelerated radiotherapy had a favorable in 20 and 13% of patients and grade 4 pneumonitis in
effect regarding time to in-field progression compared 3% of the patients. With an actuarial 2-year survival
to the standard fractionation arm (26 versus 45% at rate of 31% and a 21% local control rate, these results
Radiotherapy and Second Generation Drugs 285

compared favorably with the more toxic treatment predominantly hematological toxicity of at least grade
protocols reported so far (Pottgen et al. 2002). 4 and three cases of septic death. Esophagitis was
Kalemkerian et al. intended to determine the the main non-hematological acute toxicity. Severe
maximum tolerated dose of daily continuous intra- late side effects occurred in about 10 percent of
venous infusion of low-dose Cisplatin and Etoposide patients with pulmonary, esophageal and hematolog-
that could be administered concurrently to thoracic ical symptoms (Byhardt et al. 1995).
radiotherapy in their phase I trial published in 1999.
Continuous intravenous chemotherapy at three dose 3.2.2.3 Cisplatin/Vindesine
levels was given to 19 patients concurrently with Tsuchiya et al. (2001) reported on the JCOG 9601
radiotherapy in single daily fraction doses of 45 Gy study, a randomized phase I radiation dose-escalation
followed by a 15–20 Gy boost and three cycles trial evaluating concurrent radiochemotherapy with
of standard bolus infusion of Cisplatin 80 mg/m2 on either normo-fractionated or hyperfractionated accel-
day 1 and an Etoposide 80 mg/m2 on days 1–3. erated radiotherapy. In the normo-fractionated group
Maximum tolerated dose was determined as Cisplatin an escalation from the standard dose level of 60 Gy in
5 mg/m2/day and Etoposide 18 mg/m2/day on 5 days 30 fractions over 6 weeks to 70 Gy in 35 fractions
a week over 5 weeks. Main toxicity during concurrent over 7 weeks was planned, whereas in the hyper-
chemoradiotherapy was grade 3–4 esophagitis and fractionated accelerated arm an escalation from
myelosuppression in 42 and 47% of the patients, 54 Gy in 36 fractions over 3.6 weeks to two higher
respectively. Treatment led to a median survival time dose levels of 60 Gy in 40 fractions and 66 in 44
of 18 months with 2- and 5-year survival rates of 42 fractions was planned. Simultaneous chemotherapy
and 11%, respectively (Kalemkerian et al. 1999). consisted of two to three cycles of Cisplatin 80 mg/
Blanke et al. conducted a phase II trial including m2 on day 1 and Vindesine 3 mg/m2 on days 1 and 8
21 patients with locally advanced inoperable NSCLC every 4 weeks. 21 Patients with unresectable stage
(stage IIIA or IIIB) to assess the response rate and IIIA-B NSCLC were enrolled to the study. Treatment
toxicity of standard fractionated radiotherapy to a showed high limiting toxicity already at the first dose
dose of 50.8 Gy in 1.8 Gy per fraction, followed by a levels in 4 of 10 patients in the normo-fractionated
boost of 10 Gy in 2 Gy per fraction with concurrent arm and 7 of 11 patients in the hyperfractionated
daily low-dose Cisplatin (5 mg/m2) before each accelerated arm. Toxicity consisted mainly of hema-
fraction of radiotherapy and Etoposide (20 mg/m2) tological side effects of at least grade 3. Main
Monday to Friday on weeks 1, 2, 5, and 6. Additional non-hematologic toxicity grade 3 or greater was only
oral Etoposide (50 mg/m2) was given on the week- observed in three patients in the hyperfractionated
end. An overall response rate of 65% led to a median arm. One-year overall survival rates accounted for
overall survival of 50.2 weeks and an 1-year survival 70% in the normo-fractionated and 73% in the
rate of 45%. Therapy was associated with an high risk hyperfractionated accelerated arm, respectively. Due
of severe radiation pneumonitis, observed in 25% of to the observed high-toxicity radiation dose escalation
the patients. This was responsible for the early closure could not be performed in neither of the two groups
of the trial (Blanke et al. 1997). (Tsuchiya et al. 2001).
Long-term results of the earlier JCOG trial 8902
3.2.2.2 Cisplatin/Vinblastine were published in 2000 and showed a median 5 year
This duplet-agent regimen was investigated in the survival rate of 14.8% and a median survival of
RTOG phase II study 90-15 with hyperfractionated 14.8 months for 74 evaluated, patients with unresec-
accelerated radiation treatment to a total dose of table stage IIIA-B NSCLC. Treatment consisted of
69.9 Gy in 1.2 Gy b.i.d. combined with concur- normo-fractionated split-course radiotherapy with two
rent chemotherapy. Vinblastine 5 mg/m2 weekly was courses of 20 Gy in 2 Gy per fraction with a 14 days
given in five cycles and Cisplatin 75 mg/m2 on days break followed by another course of 10–20 Gy
1, 29 and 50. Median survival time was 12.2 months combined with simultaneous chemotherapy with
with a 1-year overall survival of 54%, estimated Cisplatin 100 mg/m2 on days 1, 29 and 57 and
2-year survival of 28% and a 1-year progression-free Vindesine 3 mg/m2 on days 1 and 8 every 4 weeks.
survival of 38%. Acute toxicity was high (45%) with Main toxicity C grade 3 was leucopenia observed in
286 M. Geier and N. Andratschke

93% of the patients. Two treatment-related deaths regimens in nonelderly patients with a 1-, 2- and
occurred and were related to pneumonia and hem- 5-year overall survival of 45, 24 and 9.1%, respec-
optysis (Kubota et al. 2000). tively and a median survival of 10 months. Local
In a French phase II study 34 patients with locally control after 5 years was 13% with a median time to
advanced NSCLC were treated with hyperfractionated local recurrence of 14 months (Jeremic et al. 1999).
accelerated radiotherapy to a total dose of 60 Gy in Another phase II trial initiated by SWOG evaluated
1.25 Gy per fraction b.i.d. with concurrent chemo- the efficacy and toxicity of Carboplatin 200 mg/m2 on
therapy consisting of daily low-dose Cisplatin 6 mg/m2 days 1 and 3 and 29 and 31 combined with intravenous
and Vindesine 2.5 mg/m2 weekly, followed by two Etoposide 50 mg/m2 on days 1 to 4 and 29 to 32
cycles of consolidating chemotherapy with Cisplatin concurrent to normo-fractionated radiotherapy of
120 mg/m2 on week 11 and 14 and vindesine 61 Gy in 1.8 to 2 Gy per fraction. Sixty-three patients
2.5 mg/m2 on weeks 11, 12 and 13. An encouraging with stage III NSCLC and considerable cardiovascu-
local failure rate of 53 and 56% at 3 and 5 years was lar, pulmonary and renal co-morbidity as well as
achieved with severe esophagitis seen in 9% of patients hearing loss, weight loss and peripheral neuropathy,
and two toxicity-related deaths. Overall 1-, 3- and which precluded Cisplatin-based treatment were
5-year survival rates of 53, 33 and 12% were observed included in the study. The treatment protocol was well
(Le Pechoux et al. 1996). tolerated in this poor performance and high-risk patient
group, with grade 3 or greater side effects of leuco-
3.2.2.4 Carboplatin/Etoposide penia, thrombocytopenia and esophagitis in 50, 23 and
Treatment regimes using Carboplatin instead of 15% of the patients. No treatment-related deaths were
Cisplatin were evaluated for poor performance observed. This very encouraging low-toxicity regimen
patients who were medically not qualified for a yielded results comparable to Cisplatin-based chemo-
treatment with Cisplatin due to co-morbidity. radiation treatment protocols in good risk patient with
Activity and feasibility of chemoradiotherapy with a 2-year overall survival rate of 21% and a median
daily low-dose carboplatin (30 mg/m2) and Etoposide overall survival of 13 months (Lau et al. 1998).
(30 mg/m2) from Monday to Friday concurrent to
hyperfractionated accelerated radiotherapy to 69.6 Gy 3.2.3 Triple Agent Regimens
in 1.2 Gy per fraction b.i.d. and increased chemother-
apy doses of 100 mg/m2, respectively on weekends 3.2.3.1 Cisplatin, Mitomycin, Vindesin
were assessed in a phase II trial by Jeremic et al.. For the A Japanese group evaluated this triple agent chemo-
41 enrolled patients with stage III NSCLC encouraging therapy scheme for concurrent chemoradiotherapy
median survival of 25 months with 3- and 5-year sur- consisting of Cisplatin 100 mg/m2 and Mitomycin
vival rates of 34 and 29%, respectively were reported 8 mg/m2 on days 1 ? 29 and Vindesine 3 mg/m2 on
by the authors, with mainly hematological (30% of days 1 ? 8 and 29 ? 36 combined with normo-
patients), esophageal (15%) and bronchopulmonary fractionated split course radiotherapy. Radiotherapy
(12%) acute side effects of at least grade 3 (Jeremic consisted of two courses of 36 Gy in 2 Gy per fraction
et al. 1998). each over 3 weeks starting on day 1 of chemotherapy
Between 1988 and 1993 58 patients older than and a rest period of 10 days between the two cycles.
70 years were enrolled in a phase II study to assess After this treatment a 2-year overall survival rate of
feasibility, toxicity and efficacy of hyperfractionated 36.7% with a median survival of 16 months was
accelerated radiotherapy to a total dose of 51 Gy in reported for the 61 eligible patients with unresectable
1.5 Gy per fraction b.i.d. concurrent to Carboplatin stage III NSCLC. Predominantly hematologic toxicity
400 mg/m2 on days 1 and 29 and oral E toposide of grade 3 or greater for leucopenia, thrombocytopenia
50 mg/m2 on days 1 to 21 and 29 to 42. Low-toxicity and anemia was observed in 95, 45 and 28% of patients,
rates were observed with grade 3 or greater hemato- respectively. Non-hematologic grade 3 or greater tox-
logical, esophageal and lung-related symptoms in 22, icity was quite low, though two treatment-related
7 and 4% of the patients without any late toxicity deaths were reported due to interstitial pneumonitis and
grade 3 or greater. Overall survival and response rates pulmonary infection after esophagobronchial fistula
were comparable to other definitive radiotherapy (Furuse et al. 1995).
Radiotherapy and Second Generation Drugs 287

From the same institution, a phase II study many phase I and II trials to evaluate toxicity and
enrolling 22 Patients with unresectable stage III efficacy in combined radiochemotherapy protocols.
NSCLC was published in 2002. Treatment included Not for all substances used unequivocal experimental
continuous radiotherapy with a total dose of 60 Gy in evidence for a true radio-sensitizing effect in the sense
2 Gy per fraction concurrent to an identical chemo- of a supra-additive effect could be demonstrated,
therapy excepting slightly dose reduced Cisplatin though. Instead, the effects may well result from
80 mg/m2 instead of 100 mg/m2 compared to the independent additive cytotoxicity and a different non-
earlier trial. The study could demonstrate a modestly overlapping toxicity profile.
improved outcome regarding survival compared to It is difficult to draw firm conclusions from these
the split-course treatment with a median survival of phase I-II trials as they significantly differed with
19 months and a similar 2 year survival rate of 34.5%. regard to fractionation, single and total dose of radio-
Furthermore lower toxicity was observed for hemato- therapy as well as the sequencing and dosage of che-
logic ([= grade 3 leucopenia and thrombocytopenia in motherapy (sequential vs. simultaneous) and the
81.8 and 26% of patients) as well as for non-hematologic combination of agents used. In addition, patient inclu-
symptoms (Atagi et al. 2002). sion criteria and tumor characteristics like stage, grade
and histology were very heterogeneous. Still, a few
3.2.3.2 Cisplatin, Mitomycin, Vinblastin interesting conclusions can be made which led to the
A South Korean group initiated a phase II study to introduction of those agents into further phase III trials.
evaluate feasibility, toxicity and treatment outcome of Radiochemotherapy in combination with second
Mitomycin, Vinblastine and Cisplatin chemotherapy generation chemotherapy agents generally proved to
concurrent to hyperfractionated accelerated radio- be feasible with acceptable, though significant toxic-
therapy in 161 stage III NSCLC patients from 1993 to ity and appeared—depending on the chemotherapy
1996. One hundred forty-six patients completed protocol—to be superior to radiotherapy alone,
treatment which consisted of Cisplatin 60 mg/m2 on especially when combined with concurrent chemo-
day 1 and 28, Vinblastine 6 mg/m2 and Mitomycin therapy. As the platinum compounds Cisplatin and
6 mg/m2 on days 2 and 29 administered simulta- Carboplatin proved to be most potent in combination
neously with radiotherapy to a total dose of 64.8 Gy with radiotherapy they formed the basis of all proto-
to 70 Gy in 1.2 Gy per fraction b.i.d.. After a mini- cols with concurrent radiotherapy either as single
mum follow-up of 45 months 1-, 2- and 5-year overall agent or as part of multiple agent protocols. Concur-
survival rates of 51.2, 25.1 and 14.8%, respectively rent radiochemotherapy protocols showed the most
and a median survival of 15 months was reported. consistent results with promising outcome data
Thrity-two patients with complete response had sig- though at the prize of considerable toxicity.
nificantly better survival (2/5 year survival 49.8/ Triple agent concurrent regimens are feasible with
39.2%) compared to patients with only partial encouraging results. Nevertheless, with considerable
response (2/5 year 22.5/11.4%). Treatment was additional toxicity the true benefit over the potent
accompanied by quite low toxicity with severe weight double agent regimen like Cisplatin/Etosposide
loss in 13.7% of patients and maximum grade 2 remains unclear.
pneumonitis in 42 Patients, although four treatment- High-dose of Ifosfamide containing induction
related deaths were reported (Lee et al. 2003). chemotherapy protocols as well as most of the
sequential chemoradiation protocols yielded disap-
pointing results both with regards to outcome and
4 Summary toxicity and therefore failed to hold to the hypothet-
ical benefit of a sequential approach. Only studies
Second generation chemotherapeutic drugs have without induction chemotherapy using HART as their
proven as a group of agents with high activity in radiotherapy regime intercalating chemotherapy in
NSCLC, particularly platinum compounds like between the split courses showed encouraging results.
Cisplatin and Carboplatin. With their mechanism of Split-course regimens and hypofractionated pro-
action and thus presumed radiation enhancing prop- tocols with insufficient biological effectiveness
erties these compounds have been incorporated in showed disappointing results, possibly not only by
288 M. Geier and N. Andratschke

counteracting the effect of radiotherapy, but also by dose cisplatin and etoposide with concurrent thoracic
obscuring a possible beneficial effect of the chemo- radiotherapy in unresectable stage III non-small-cell lung
cancer. Int J Radiat Oncol Biol Phys 37:111–116. doi:
therapy regime. S0360301696004786
The results of low-dose platinum-based concurrent Boven E, Tierie AH, Stam J, Pinedo HM (1988) Combined
radiochemotherapy regimens are very intriguing, radiation therapy and daily low-dose cisplatin for inoper-
though whether the interaction is through synergistic able, locally advanced non-small-cell lung cancer: results of
a phase II trial. Semin Oncol 15:18–19
radio-sensitizing effects or rather reflects independent, Byhardt RW, Scott CB, Ettinger DS, Curran WJ, Doggett RL,
additive cytotoxicity due to cumulative toxic effects is Coughlin C, Scarantino C, Rotman M, Emami B (1995)
still a matter of debate. Non-hematologic toxicity, Concurrent hyperfractionated irradiation and chemotherapy
particularly esophageal, became only relevant, when a for unresectable non-small-cell lung cancer. Results of
radiation therapy oncology group 90–15. Cancer
second chemotherapeutic agent was added to the 75:2337–2344
concurrent regimen. Chen AY, Okunieff P, Pommier Y, Mitchell JB (1997)
Despite the heterogeneity of the studies, radio- Mammalian DNA topoisomerase I mediates the enhance-
chemotherapy with second generation chemotherapy ment of radiation cytotoxicity by camptothecin derivatives.
Cancer Res 57:1529–1536
proved to be feasible and—especially concurrent Dewit L (1987) Combined treatment of radiation and cis-
double agent platinum-based protocols—appeared to diamminedichloroplatinum (II): a review of experimental
be superior to conventional radiotherapy alone and and clinical data. Int J Radiat Oncol Biol Phys 13:403–426
has been considered a value treatment option in Douple EB, Richmond RC (1978) Platinum complexes as
radiosensitizers of hypoxic mammalian cells. Br J Cancer
inoperable advanced stage NSCLC worthwhile testing Suppl 3:98–102
in subsequent phase III trials. Fleming RA (1997) An overview of cyclophosphamide and
ifosfamide pharmacology. Pharmacotherapy 17:146S–154S
Furuse K, Kubota K, Kawahara M, Kodama N, Ogawara M,
Akira M, Nakajima S, Takada M, Kusunoki Y, Negoro S
References et al (1995) Phase II study of concurrent radiotherapy and
chemotherapy for unresectable stage III non-small-cell lung
cancer. Southern Osaka lung cancer study group. J Clin
Albain KS, Crowley JJ, Turrisi AT 3rd, Gandara DR, Oncol 13:869–875
Farrar WB, Clark JI, Beasley KR, Livingston RB (2002) Grau C, Overgaard J (1991) Radiosensitizing and cytotoxic
Concurrent cisplatin, etoposide, and chest radiotherapy in properties of mitomycin C in a C3H mouse mammary
pathologic stage IIIB non-small-cell lung cancer: a South- carcinoma in vivo. Int J Radiat Oncol Biol Phys
west Oncology Group phase II study, SWOG 9019. J Clin 20:265–269
Oncol 20:3454–3460 Hazuka MB, Crowley JJ, Bunn PA Jr, O’Rourke M, Braun TJ,
Ardizzoni A, Grossi F, Scolaro T, Giudici S, Foppiano F, Boni Livingston RB (1994) Daily low-dose cisplatin plus
L, Tixi L, Cosso M, Mereu C, Ratto GB, Vitale V, Rosso R concurrent high-dose thoracic irradiation in locally
(1999) Induction chemotherapy followed by concurrent advanced unresectable non-small-cell lung cancer: results
standard radiotherapy and daily low-dose cisplatin in locally of a phase II Southwest Oncology group study. J Clin Oncol
advanced non-small-cell lung cancer. Br J Cancer 12:1814–1820
81:310–315. doi:10.1038/sj.bjc.6690693 Jeremic B, Shibamoto Y, Milicic B, Nikolic N, Dagovic A,
Atagi S, Kawahara M, Hosoe S, Ogawara M, Kawaguchi T, Milisavljevic S (1998) Concurrent radiochemotherapy for
Okishio K, Naka N, Sunami T, Mitsuoka S, Akira M (2002) A patients with stage III non-small-cell lung cancer (NSCLC):
phase II study of continuous concurrent thoracic radiotherapy long-term results of a phase II study. Int J Radiat Oncol Biol
in combination with mitomycin, vindesine and cisplatin in Phys 42:1091–1096. doi:S0360-3016(98)00283-1[pii]
unresectable stage III non-small-cell lung cancer. Lung Jordan MA, Wilson L (2004) Microtubules as a target for
Cancer 36:105–111. doi:S0169500201004603 anticancer drugs. Nat Rev Cancer 4:253–265. doi:10.1038/
Bardet E, Riviere A, Charloux A, Spaeth D, Ducolone A, Le nrc131710.1038/nrc1317
Groumellec A, Pellae-Cosset B, Henry-Amar M, Douillard Jeremic B, Shibamoto Y, Milicic B, Milisavljevic S, Nikolic N,
JY (1997) A phase II trial of radiochemotherapy with daily Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999) A
carboplatin, after induction chemotherapy (carboplatin and phase II study of concurrent accelerated hyperfractionated
etoposide), in locally advanced non-small-cell lung cancer: radiotherapy and carboplatin/oral etoposide for elderly
final analysis. Int J Radiat Oncol Biol Phys 38:163–168. doi: patients with stage III non-small-cell lung cancer. Int J Radiat
S036030169700254X Oncol Biol Phys 44:343–348. doi:S0360-3016(99)00006-1
Binaschi M, Zunino F, Capranico G (1995) Mechanism of Kalemkerian GP, Belzer K, Wozniak AJ, Gaspar LE,
action of DNA topoisomerase inhibitors. Stem Cells Valdivieso M, Kraut MJ (1999) Phase I trial of concurrent
13:369–379. doi:10.1002/stem.5530130408 thoracic radiation and continuous infusion cisplatin and
Blanke C, DeVore R, Shyr Y, Epstein B, Murray M, Hande K, etoposide in stage III non-small-cell lung cancer. Lung
Stewart S, Johnson D (1997) A pilot study of protracted low Cancer 25:175–182. doi:S0169500299000604
Radiotherapy and Second Generation Drugs 289

Kelly K, Hazuka M, Pan Z, Murphy J, Caskey J, Leonard C, conformal radiotherapy and concurrent mitomycin-C,
Bunn PA Jr (1998) A phase I study of daily carboplatin and vinblastine, and cisplatin chemotherapy for Stage III locally
simultaneous accelerated, hyperfractionated chest irradia- advanced, unresectable, non-small-cell lung cancer. Int J
tion in patients with regionally inoperable non-small-cell Radiat Oncol Biol Phys 56:996–1004. doi:S03603016
lung cancer. Int J Radiat Oncol Biol Phys 40:559–567. doi: 03001275
S0360301697007694 Mirimanoff RO, Moro D, Bolla M, Michel G, Brambilla C,
Komaki R, Scott C, Ettinger D, Lee JS, Fossella FV, Curran W, Mermillod B, Miralbell R, Alberto P (1998) Alternating
Evans RF, Rubin P, Byhardt RW (1997) Randomized study radiotherapy and chemotherapy for inoperable Stage III
of chemotherapy/radiation therapy combinations for favor- non-small-cell lung cancer: long-term results of two Phase
able patients with locally advanced inoperable non-small- II GOTHA trials. Groupe d’Oncologie Thoracique Alpine.
cell lung cancer: radiation therapy oncology group (RTOG) Int J Radiat Oncol Biol Phys 42:487–494. doi:S036030
92–04. Int J Radiat Oncol Biol Phys 38:149–155. doi: 1698002466
S0360-3016(97)00251-4 Palazzi M, Morrica B, Montanaro P, Cerizza L, Leoni M (1996)
Komaki R, Seiferheld W, Ettinger D, Lee JS, Movsas B, Sause Hyperfractionated radiotherapy and concomitant cisplatin in
W (2002) Randomized phase II chemotherapy and radio- stage III non-small cell lung cancer: a phase II study by
therapy trial for patients with locally advanced inoperable the AIRO-Lombardia Cooperative Group. Lung Cancer 15:
non-small-cell lung cancer: long-term follow-up of RTOG 85–91. doi:0169500296005739
92–04. Int J Radiat Oncol Biol Phys 53:548–557. doi: Planting A, Helle P, Drings P, Dalesio O, Kirkpatrick A, McVie
S0360301602027931 G, Giaccone G (1996) A randomized study of high-dose
Kubota K, Tamura T, Fukuoka M, Furuse K, Ikegami H, split course radiotherapy preceded by high-dose chemo-
Ariyoshi Y, Kurita Y, Saijo N (2000) Phase II study of therapy versus high-dose radiotherapy only in locally
concurrent chemotherapy and radiotherapy for unresectable advanced non-small-cell lung cancer. An EORTC Lung
stage III non-small-cell lung cancer: long-term follow-up Cancer Cooperative Group trial. Ann Oncol 7:139–144
results. Japan clinical oncology group protocol 8902. Ann Pottgen C, Eberhardt W, Bildat S, Stuben G, Stamatis G,
Oncol 11:445–450 Hillejan L, Sohrab S, Teschler H, Seeber S, Sack H,
Latz D, Weber KJ (2002) Transient DNA double-strand Stuschke M (2002) Induction chemotherapy followed by
breakage in 4-hydroperoxyifosfamide-treated mammalian concurrent chemotherapy and definitive high-dose radio-
cells in vitro does not interact with the rejoining of therapy for patients with locally advanced non-small-cell
radiation-induced double-strand breaks. Strahlenther Onkol lung cancer (stages IIIa/IIIb): a pilot phase I/II trial. Ann
178:269–274 Oncol 13:403–411
Latz D, Schulze T, Manegold C, Schraube P, Flentje M, Weber Pujol JL, Lafontaine T, Quantin X, Reme-Saumon M, Cupissol
KJ (1998) Combined effects of ionizing radiation and 4- D, Khial F, Michel FB (1999) Neoadjuvant etoposide,
hydroperoxyfosfamide in vitro. Radiother Oncol ifosfamide, and cisplatin followed by concomitant thoracic
46:279–283. doi:S0167814097001941 radiotherapy and continuous cisplatin infusion in stage IIIb
Lau DH, Crowley JJ, Gandara DR, Hazuka MB, Albain KS, non-small cell lung cancer. Chest 115:144–150
Leigh B, Fletcher WS, Lanier KS, Keiser WL, Livingston Sarihan S, Darendeliler E, Kizir A, Tuncel N, Oral EN,
RB (1998) Southwest oncology group phase II trial of Karadeniz A, Bilge N (1998) A phase II trial, feasibility of
concurrent carboplatin, etoposide, and radiation for poor- combination of daily cisplatinum and accelerated radiother-
risk stage III non-small-cell lung cancer. J Clin Oncol apy via concomitant boost in stage III non-small cell lung
16:3078–3081 cancer. Lung Cancer 20:37–46. doi:S0169500298000038
Le Pechoux C, Arriagada R, Le Chevalier T, Bretel JJ, Cosset Scagliotti GV, Ricardi U, Crino L, Maranzano E, De Marinis F,
BP, Ruffie P, Baldeyrou P, Grunenwald D (1996) Concur- Morandi MG, Meacci L, Marangolo M, Emiliani E, Rosti G,
rent cisplatin-vindesine and hyperfractionated thoracic Figoli F, Bolzicco G, Masiero P, Gentile A, Tonato M
radiotherapy in locally advanced non-small-cell-lung can- (1996) Phase II study of intensive chemotherapy with
cer. Int J Radiat Oncol Biol Phys 35:519–525. doi: carboplatin, ifosfamide and etoposide plus recombinant
0360301696001484 human granulocyte colony-stimulating factor and sequential
Lee JS, Scott C, Komaki R, Fossella FV, Dundas GS, radiotherapy in locally advanced, unresectable non-small-
McDonald S, Byhardt RW, Curran WJ Jr (1996) Concurrent cell lung cancer. Cancer Chemother Pharmacol 38:561–565
chemoradiation therapy with oral etoposide and cisplatin for Sekine I, Nishiwaki Y, Ogino T, Yokoyama A, Saito M, Mori
locally advanced inoperable non-small-cell lung cancer: K, Tsukiyama I, Tsuchiya S, Hayakawa K, Yoshimura K,
radiation therapy oncology group protocol 91–06. J Clin Ishizuka N, Saijo N (2002) Phase II study of twice-daily
Oncol 14:1055–1064 high-dose thoracic radiotherapy alternating with cisplatin
Lee JS, Komaki R, Fossella FV, Glisson BS, Hong WK, Cox and vindesine for unresectable stage III non-small-cell lung
JD (1998) A pilot trial of hyperfractionated thoracic cancer: Japan Clinical Oncology Group Study 9306. J Clin
radiation therapy with concurrent cisplatin and oral etopo- Oncol 20:797–803
side for locally advanced inoperable non-small-cell lung Sui M, Fan W (2005) Combination of gamma-radiation
cancer: a 5 year follow-up report. Int J Radiat Oncol Biol antagonizes the cytotoxic effects of vincristine and vinblas-
Phys 42:479–486. doi:S0360-3016(98)00247-8 tine on both mitotic arrest and apoptosis. Int J Radiat Oncol
Lee SW, Choi EK, Lee JS, Lee SD, Suh C, Kim SW, Kim WS, Biol Phys 61:1151–1158. doi:S0360-3016(04)03144-X [pii]
Ahn SD, Yi BY, Kim JH, Noh YJ, Kim SS, Koh Y, Kim Thomas P, Kleisbauer JP, Robinet G, Clavier J, Poirier R,
DS, Kim WD (2003) Phase II study of three-dimensional Vernenegre A, Bonnaud F, Taytard A, Paillotin D, Pommier
290 M. Geier and N. Andratschke

De Santi P, Barriere JR, Pignon T (1997) Carboplatin as Van Belle S, Fortan L, De Smet M, De Neve W, Van der Elst J,
radiosensitizer in non-small cell lung cancer after cisplatin Storme G (1994) Interaction between vinblastine and
containing chemotherapy. A phase I study of a groupe ionizing radiation in the mouse MO4 fibrosarcoma in vivo.
francais de pneumo-cancerologie (G.F.P.C.). Lung Cancer Anticancer Res 14:1043–1048
18:71–81. doi:S0169-5002(97)00047-0 Van den Brande P, De Ruysscher D, Vansteenkiste J, Spaas P,
Tsuchiya S, Ohe Y, Sugiura T, Fuwa N, Kitamoto Y, Mori K, Specenier P, Demedts M (1998) Sequential treatment with
Kobayashi H, Nakata K, Sawa T, Hirai K, Etoh T, Saka H, vindesine-ifosfamide-platinum (VIP) chemotherapy fol-
Saito A, Fukuda H, Ishizuka N, Saijo N (2001) Randomized lowed by platinum sensitized radiotherapy in stage IIIB
phase I study of standard-fractionated or accelerated- non-small cell lung cancer: a phase II trial. Lung Cancer
hyperfractionated radiotherapy with concurrent cisplatin 22:45–53. doi:S0169-5002(98)00071-3
and vindesine for unresectable non-small cell lung cancer: a van Harskamp G, Boven E, Vermorken JB, van Deutekom H,
report of Japan Clinical Oncology Group Study (JCOG Stam J, Njo KH, Karim AB, Tierie AH, Golding RP, Pinedo
9601). Jpn J Clin Oncol 31:488–494 HM (1987) Phase II trial of combined radiotherapy and
Uitterhoeve AL, Belderbos JS, Koolen MG, van der Vaart daily low-dose cisplatin for inoperable, locally advanced
PJ, Rodrigus PT, Benraadt J, Koning CC, Gonzalez non-small cell lung cancer (NSCLC). Int J Radiat Oncol
Gonzalez D, Bartelink H (2000) Toxicity of high-dose Biol Phys 13:1735–1738
radiotherapy combined with daily cisplatin in non-small Verweij J, Pinedo HM (1990) Mitomycin C: mechanism of
cell lung cancer: results of the EORTC 08912 phase I/II action, usefulness and limitations. Anticancer Drugs 1:5–13
study. European Organization for Research and Ziegler W, Kopp JM (1987) The effect of combined treatment of
Treatment of Cancer. Eur J Cancer 36:592–600. doi: HeLa cells with cisplatin and irradiation upon survival and
S0959804999003159 recovery from radiation damage. Radiother Oncol 8:71–78
Radiotherapy and Third Generation
Concurrent Chemotherapy Agents
Ross Bland, Puneeth Iyengar, and Hak Choy

Contents Abstract
Evidence shows that treating locally advanced
1 Carboplatin and Paclitaxel..................................... 292 non-small cell lung cancer (NSCLC) patients with
2 Vinorelbine and Gemcitabine ................................ 297 concurrent radiation and second generation chemo-
therapy agents provides a survival benefit when
3 Docetaxel................................................................... 302
compared to delivering the same therapy sequen-
4 Irinotecan.................................................................. 303 tially. However, survival results remain dismal and
5 Pemetrexed ............................................................... 304 concurrent chemoradiation exhibits significant tox-
6 Third Generation Agents and Dose Escalation ... 305
icity. As third generation chemotherapy agents have
become available, their radiation-sensitizing proper-
References.......................................................................... 305
ties have been extensively studied in the laboratory.
Subsequently, clinical studies have been performed
to evaluate the potential benefit of incorporating
these newer agents in concurrent chemoradiation
regimens. This chapter aims to provide insight into
how these agents were transitioned from the labora-
tory bench to their current role in the standard of care
of patients with locally advanced disease.

Despite advances in the treatment of many cancers


over the last three decades, our ability to control
stage III lung cancer continues to be poor. A majority
of these lesions are not candidates for surgical
resection and are treated with a combination of che-
motherapy, biologic agents, and external beam radi-
ation. As part of concurrent therapy, systemic agents
primarily play a role in potentially sensitizing tumor
cells to the effects of ionizing radiation. They also
serve to attempt to control disseminated disease.
The original platinum-based drug, cisplatin, as
R. Bland  P. Iyengar  H. Choy (&) well as topoisomerase targeted systemic agents have
Department of Radiation Oncology, had a unique capacity to synergize with ionizing
UT Southwestern Medical Center at Dallas,
Dallas, USA radiation in promoting lung tumor cell death. How-
e-mail: Hak.Choy@utsouthwestern.edu ever, survival outcomes with these agents have not

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 291
DOI: 10.1007/174_2011_269, Ó Springer-Verlag Berlin Heidelberg 2011
292 R. Bland et al.

been good enough and normal tissue toxicities have of polymers of tubulin in dynamic equilibrium with
been excessive. Therefore, there has been a great tubulin heterodimers. In contrast to vinca alkaloids,
effort over recent years to provide robust pre-clinical which promote disassembly of microtubules, paclitaxel
evidence of the potential of new systemic agents to be acts in the opposite direction to induce polymerization
used concurrently with ionizing radiation in the (Rowinsky and Donehower 1995). In the presence of
treatment of lung cancer. Slowly, but surely, this paclitaxel, these abnormally stable microtubules are
preclinical work has translated into early and dysfunctional and the dividing cell is halted in the G2/
advanced phase clinical trials that have offered hope M phase of the cell cycle (Schiff et al. 1979; Parness
regarding a new generation of therapies. and Horwitz 1981; Manfredi et al. 1982). Using ham-
This chapter will summarize some of these new ster cells in culture, Sinclair and Morton demonstrated
advances, from rationale and preclinical data to find- that cells in the M phase were most radiation sensitive
ings in the latest clinical trials. We hope to offer some while those in early G1 and late S phases were the
insight into how these agents work with radiation in most resistant. The ratio of colony-surviving fractions
promoting tumor kill under various circumstances. We for these two cell populations was almost fourfold
will also attempt to offer some hope regarding newer (Sinclair and Morton 1966). In a study using the human
classes of cytotoxic chemotherapeutics and their syn- leukemic cell line, HL-60, up to 70% of cells were
ergy with radiation. The format of the chapter hopefully blocked in the G2/M phase after exposure to low-dose
allows the reader the opportunity to appreciate the paclitaxel (30 nmol/L) for 1 h. These paclitaxel-
evolution of third generation chemotherapies given exposed cells were also treated with radiation doses
concurrently with radiation therapy from preclinical ranging from 0 to 4 Gy at 24 h and the sensitizing
data to altering practice patterns and standards of enhancement ratio of paclitaxel was reported to be 1.48
care. Some of the concurrent systemic agents we (Choy et al. 1993). Several studies showed that pac-
highlight and focus on include carboplatin, paclitaxel, litaxel could provide radiation sensitization in the rel-
vinorelbine, gemcitabine, docetaxel, and pemetrexed. atively radiation resistant human astrocytoma cell line
A number of these agents have begun to represent part and ovarian cell lines. In these studies, the concentra-
of the standard of care for treatment of lung cancer in tion of paclitaxel and the fraction of cells in the G2 and
the definitive setting, while others have been too toxic M phases of the cell cycle determined the degree of
and/or less than efficacious. enhancement. Paclitaxel exhibited an enhancement
ratio of 1.8 at 10 and 1.2 at 1 mmol/l (Tishler et al.
1992a, b; Stern et al. 1993). Researchers took advan-
1 Carboplatin and Paclitaxel tage of the synergy of action within the cell cycle when
utilizing paclitaxel and radiation in combined modality
A meta-analysis of all randomized trials from the therapy, especially in the setting of ordinarily radiation
pretaxane era showed that adding cisplatin-based che- resistant tumor systems.
motherapy to radiotherapy for locally advanced Based on activity in advanced disease and preclin-
NSCLC provided a 13% reduction in the risk of death. ical data indicating impressive radiation enhancement
However, the absolute survival advantage was a of paclitaxel in various cell lines, a phase I trial of
modest 4% at 2 years and 2% at 5 years (NSCLC Coll paclitaxel with concurrent radiotherapy for inoperable
Group 1995). The results of this meta-analysis dem- NSCLC was initiated (Choy et al. 1994). This study
onstrated the benefit of chemotherapy but also made demonstrated that thoracic radiotherapy could be safely
clear the need for improved treatment regimens. administered concurrently with weekly 3 h infusions of
Meanwhile, there was a growing interest in paclitaxel, paclitaxel at 60 mg/m2 for six weeks. Esophagitis was
a chemotherapy agent first approved for use in the the dose limiting toxicity. Notably, this was a lower but
palliative therapy of patients with ovarian and breast more frequent dose when compared to the every-
cancers resistant to conventional chemotherapy. 3-week paclitaxel regimen used in phase II studies of
Paclitaxel’s mechanism of action involves regulation paclitaxel alone. Subsequently, a phase II study of
of microtubule formation. Properly functioning weekly paclitaxel and concurrent radiation for locally
microtubules are required for formation of the mitotic advanced NSCLC (Choy and Safran 1995) was con-
spindle during cell division. Microtubules are made up ducted and showed encouraging response and survival
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 293

rates when compared to the most active cisplatin-based This regimen resulted in an overall response rate of
chemoradiation regimens (Dillman et al. 1990, 1996; 76%, a one year survival rate of 56%, and acceptable
Lee et al. 1996; Schaake-Koning et al. 1992; NSCLC toxicity (Choy et al. 1998). In a phase II trial evaluating
Coll Group 1995). weekly carboplatin (100 mg/m2) and paclitaxel
Carboplatin, a cisplatin analog, was known to be (45 mg/m2) with concurrent RT (60-65 Gy), Belani
less nephrotoxic and emetogenic than cisplatin but et al. reported 1- and 3-year survival rates of 63 and
have similar antitumor activity (Rowinsky et al. 1993; 54%, respectively (Belani et al. 1997). The California
Klastersky et al. 1990, 1993; Kosmidis et al. 1997). Cancer Consortium reasoned that more frequent
Data from preclinical studies indicated that carbo- administration of paclitaxel should optimize its radia-
platin enhances ionizing radiation through such tion sensitizing effect. This led to a phase II trial to
mechanisms as radiosensitization of hypoxic cells, evaluate the safety and efficacy of twice-weekly
inhibition of the repair of sublethal or potentially paclitaxel (30 mg/m2), weekly carboplatin (AUC
lethal damage, binding to thiols, and increased 1.5 mg/mL 9 min), and concurrent radiation therapy
induction of chromosomal aberrations (Begg et al. (61 Gy) followed by consolidation with two 21-day
1987, 1989; Coughlin and Richmond 1989; Douple cycles of carboplatin (AUC 6 mg/mL 9 min) and
1993). These findings suggested that, at least in vitro, paclitaxel (200 mg/m2). This treatment regimen
radiation enhancement by carboplatin is similar to resulted in a response rate of 71% and a median survival
that exhibited by cisplatin. However, since compared time of 17 months and was deemed tolerable by the
to cisplatin, carboplatin could provide a greater investigators (Lau et al. 2001).
intracellular platinum concentration during irradia- While studies showed that reduced dose carbo-
tion, it was thought that carboplatin may result in platin and paclitaxel with concurrent radiation was
more effective synergy with radiation (Belani 1993; effective and tolerable for locally advanced NSCLC,
Micetich et al. 1985). A phase II study examined the benefit of either induction or consolidation full-
carboplatin 75 mg/m2 weekly in combination with dose chemotherapy in conjunction with concurrent
thoracic radiation. Overall the treatment was well chemoradiotherapy was uncertain. A phase II ran-
tolerated and showed a 1-year survival rate of 50% in domized three arm study was undertaken to address
stage III NSCLC patients (Clark et al. 1998). this uncertainty (Belani et al. 2005). Arm 1, the
Laboratory data suggested a synergistic effect sequential arm, delivered two cycles of induction
when combining the two systemic antineoplastics carboplatin and paclitaxel followed by radiation.
carboplatin and paclitaxel (Bunn and Kelly 1995). Arm 2, the induction/concurrent arm, delivered two
In initial phase I and II trials, combination regimens cycles of induction carboplatin and paclitaxel fol-
of carboplatin and paclitaxel used to treat advanced lowed by weekly carboplatin and paclitaxel with
(stages III and IV) NSCLC exhibited overall response concurrent radiation. Finally, arm 3, the concurrent/
rates as high as 63% (Langer et al. 1995; Johnson consolidation arm consisted of weekly carboplatin and
1999; Johnson et al. 1996; Vafai et al. 1995). Fur- paclitaxel with concurrent radiation followed by two
thermore, these studies demonstrated that combined cycles of consolidation carboplatin and paclitaxel. All
carboplatin and paclitaxel regimens for advanced three treatment regimens were felt to be safe with
NSCLC had manageable toxicities. Based on the median survival times of 13, 12.7, and 16.3 months in
success of these agents in advanced disease and their arms 1, 2, and 3, respectively. As expected, the two
known radiation enhancing properties, studies were arms involving concurrent chemoradiation exhibited
conducted to evaluate the activity and toxicity of the greatest toxicity. Esophagitis was seen in 3% of
carboplatin and paclitaxel with concurrent radiation patients in arm 1, 19% of patients in arm 2, and 28% of
therapy in patients with locally advanced NSCLC patients in arm 3. Lung toxicities were identified in 7%
(see Table 1). A multi-institutional phase II trial of arm 1 patients, 4% of arm 2 patients, and 16% of
administered weekly regimens of carboplatin (AUC arm 3 patients. Although there was not a statistically
2 mg/mL 9 min) and paclitaxel (50 mg/m2) with significant difference in survival between arms, the
concurrent radiation therapy (66 Gy in 33 fractions) results suggested an improved outcome with con
followed by consolidation carboplatin (AUC 6 mg/mL current chemoradiation followed by consolidation
9 min) and paclitaxel (200 mg/m2) every three weeks. chemotherapy.
Table 1 Summary of selected paclitaxel chemoradiation studies
294

Reference Study Number of Ind Conc Cons MST Comments


patients (months)
Choy et al. (1998) Phase II 40 – Carbo/Pac/66 Gy Carbo/Pac 20.5 Regimen appears safe and active.
Belani et al. (1997) Phase II 38 – Carbo/Pac/60-65 Gy – 63% Regimen appears safe and active.
(1 year)
Ratanatharathorn Phase II 30 – Carbo/Pac/60 Gy Carbo/Pac 14.5 Effective and tolerable regimen.
et al. (2001)
Lau et al. (2001) Phase II 34 – Carbo/Pac(2/ Carbo/Pac 17 Concurrent twice weekly Pac with
week)/61 Gy weekly Carbo is safe and active.
Solomon et al. Phase I/II 25 – P/Pac(2/week)/60 Gy Carbo/Pac 23.6 Concurrent twice weekly Pac with
(2003) weekly P is safe and active.
Kaplan et al. (2004) Phase II 19 – Carbo/Pac/66 Gy Carbo/Pac 13.9 Regimen is feasible and well tolerated.
Belani et al. (2005) LAMP, Phase II 257 Carbo/Pac 63 Gy – 13 Arm 3 was numerically most efficacious
Carbo/Pac Carbo/Pac/63 Gy – 12.7 but most toxic.

– Carbo/Pac/63 Gy Carbo/Pac 16.3


Akerley et al. CALGB 9534, Phase 40 Carbo/Pac Carbo/Pac/66 Gy – 27% Included patients with [5% weight
(2005) II (3 years) loss, but survival consistent with more
selective studies.
Divers et al. (2005) Phase I/IIa 35 P/Gem Gem/Pac q – 17 Regimen is safe and effective in good
3 weeks/60 Gy performance status patients.
Huber et al. (2006) CTRT99/97 214 Carbo/Pac 60 Gy – 14.7 Concurrent chemoradiation arm had
BROCAT, Phase III Carbo/Pac Pac/60 Gy – 18.7 improved survival.

Davies et al. (2006) S9712 54 – Carbo/E/61 Gy Pac 10.2 Compared to S429, in poor risk patients,
Pac consolidation does not increase
survival.
Vokes et al. (2007) CALGB 39801, 366 – Carbo/Pac/66 Gy – 12 Induction CT increased toxicity without
Phase II Carbo/Pac Carbo/Pac/66 Gy – 14 survival benefit. Low MSTs possibly
due to low dose 3rd generation CT.
(continued)
R. Bland et al.
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 295

MST median survival time, LAMP locally advanced multi-modality protocol, CALGB Cancer and Leukemia Group B, WJTOG West Japan Thoracic Oncology Group, Carbo
The first phase III trial to evaluate concurrent

Arm 3 was equally as efficacious but


carboplatin and paclitaxel with radiation therapy for

esophagitis but SS improvement in


With AM, no SS reduction in locally advanced NSCLC was conducted by the
Cancer and Leukemia Group B (CALGB) (Vokes

less toxic than other arms.


patient self-assessments. et al. 2007). CALGB 39801 was designed to test the
value of induction chemotherapy in the context of
concurrent chemoradiotherapy. Patients were ran-
domized to carboplatin and paclitaxel with concurrent
radiotherapy (concurrent arm), or to induction car-
Comments

boplatin and paclitaxel followed by carboplatin and


paclitaxel based concurrent chemoradiotherapy
(induction/concurrent arm). There was no statistically
significant difference in survival. Median survival was
(months)

12 months on the concurrent arm versus 14 months


carboplatin, Pac paclitaxel, MVP mitomycin/vindesine/cisplatin, AM amofostine, CT chemotherapy, SS statistically significant
MST

17.3

17.9

19.8
20.5

on the induction/concurrent arm. Additionally, the


22

induction/concurrent arm demonstrated increased


Carbo/Pac

neutropenia and overall maximal toxicity. This study


Irinotecan
Carbo/

failed to show a benefit for the role of induction


MVP
Cons

chemotherapy in the context of concurrent chemora-


diotherapy analogous to what was reported by Belani


Carbo/Irinotecan/60 Gy

et al. (2005). One possible explanation for the poor


Carbo/Pac/69.6 Gy,

Carbo/Pac/69.6 Gy,

performance of the concurrent arm, is that the patients


MVP/60 Gy (split)

Carbo/Pac/60 Gy

on this arm never received systemic full dose che-


1.2 Gy/bid AM

motherapy. The radiation-sensitizing dose was given


1.2 Gy bid

concurrently with radiation was not optimal for


Conc

treating micrometastatic disease.


Studies of concurrent chemoradiation demon-
strated improved survival at the cost of increased
Carbo/Pac

Carbo/Pac

toxicity, particularly esophagitis, when compared to


sequential chemoradiation (Komaki et al. 2003).
Ind


Movsas et al. performed a quality-adjusted survival


(QAS) analysis of several RTOG lung cancer studies
Number of

and found that the QAS of sequential chemoradiation


patients

was nearly equivalent to that of concurrent chemo-


243

456

radiation (Movsas et al. 1999). Reduction in esopha-


geal, upper GI and lung toxicities was associated with
the greatest improvement in QAS. In light of these
WJTOG0105, Phase
RTOG 9801, Phase

findings, RTOG 98-01 was designed to test amofos-


tine’s (AM) ability to reduce the severity of esopha-
gitis in concurrent chemoradiation regimens (Movsas
et al. 2003). AM is a cytoprotectant that is dephos-
Study

phorylated in tissue and subsequently acts as a scav-


III

III

enger of radiation induced oxygen free radicals


Table 1 (continued)

Movsas et al. (2003)

(Calabro-Jones et al. 1985; Wasserman and Chapman


Yamamoto et al.

2004). In RTOG 98-01, 243 patients with locally


advanced NSCLC were administered induction car-
Reference

boplatin and paclitaxel followed by hyperfractionated


(2010)

radiotherapy (69.6 at 1.2 Gy bid) with concurrent


carboplatin and paclitaxel. Patients were randomized
296

Table 2 Summary of selected vinorelbine or gemcitabine chemoradiation studies


Reference Study Patients Induction Concurrent Consolidation MST Comments
(months)
Vokes et al. (2002) CALGB 9431, Phase II 175 P/Gem P/Gem/66 Gy – 18.3 Greater MSTs than in CALGB trials
P/Pac P/Pac/66 Gy – 14.8 using squential CTRT.

P/Vrb P/Vrb/66 Gy – 17.7


Zatloukal et al. Czech Republic 102 P/Vrb P/Vrb/60 Gy – 16.6 SS survival benefit to concurrent CTRT.
(2004) P/Vrb 60 Gy – 12.9
Lee et al. (2005) Phase II 40 Gem/Vrb P/E/63 Gy – 23.2 Regimen had promising survival results.
Fournel et al. GFPC-GLOT-IFCT 02-01, 133 P/Pac P/Vrb/66 Gy – 19.3 Similar toxicities but numercially
(2006) Phase II – P/Vrb/66 Gy P/Pac 16.9 greater MST in arm 1.

Kim et al. (2007) Phase III 134 P/Gem P/Pac/66 Gy – 12.6 Induction arm had SS worse PFS.
– P/Pac/66 Gy – 18.2
Rusu et al. (2007) Phase II 57 – Platinum/Vrb/RT Platinum/Vrb 15 Regimen is active and well tolerated.
Hirsh et al. (2007) Phase II 41 Carbo/ Gem/Pac/60 Gy – 25 Regimen is effective and well tolerated.
Gem
Krzakowski et al. Phase II 54 PO Vrb/ PO Vrb/P/66 Gy – 23.4 With oral Vrb, high rate of treatment
(2008) P completion. Phase III trial with oral Vrb
underway.
Choy et al. (2009) RTOG 0017, Phase I 27 – Carbo/Gem/63 Gy Carbo/Gem 13.3 Lowest dose Gem/Pac too toxic with
8 – Gem/Pac/63 Gy Carbo/Gem 12.9 RT. Established MTD of Carbo/Gem
with RT.
Blanco et al. (2008) ACROSS, Phase II 44 P/Gem Gem/68.4 Gy – 17.7 Regimen had substantial toxicity.
Leong et al. (2010) Phase II 42 Gem/Vrb Vrb/60-66 Gy – 17 This non-platinum regimen is feasible,
tolerable, and effective.
MST median survival time, CALGB Cancer and Leukemia Group B, CTRT chemoradiation, GFPC French Pneumonology Group, GLOT Groupe Lyon-Saint-Etienne d’Oncologie
Thoracique, IFCT Intergroupe Francophone de Cancérologie Thoracique, Carbo carboplatin, Pac paclitaxel, Vrb vinorelbine, P cisplatin, Gem gemcitabine, CTRT chemora-
diation, RT radiation therapy, CT chemotherapy, SS statistically significant, MTD maximum tolerated dose, PFS progression free survival
R. Bland et al.
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 297

to receive AM 500 mg IV four times per week or no carboplatin and paclitaxel with concurrent radiation
AM during chemoradiation. The rate of grade 3 or followed by carboplatin and paclitaxel consolidation.
worse esophagitis was not significantly different The median survival times for the three study arms
between arms (AM, 30 versus no AM, 34%). How- were not significantly different (arm A: 20, arm B:
ever, patient diaries reported significantly lower 19.8, arm C: 22 months). Unfortunately, the study
swallowing dysfunction with AM and the quality of was not able to demonstrate non-inferiority. While
life instrument (EORTC QLQ) demonstrated a sig- difference in survival for the three arms was not sig-
nificant difference in pain that favored the AM arm. nificant, hematologic and gastrointestinal toxicities
Median survival rates were similar (AM, 17.3 versus were more pronounced in the control arm. Other than
no AM, 17.9 months). Thus, while NCI-CTC toxicity neurotoxicity, most of the toxicities were the mildest
endpoints did not support the hypothesis that AM in the carboplatin and paclitaxel arm. Based on these
given during concurrent chemoradiation reduces data of efficacy and toxicity, the WJTOG selected the
esophagitis, the patient-derived self-assessments carboplatin and paclitaxel study arm as the reference
suggested a possible benefit from AM. This discon- treatment regimen for future phase III studies of
nect between the patient’s evaluation of difficulty locally advanced NSCLC.
swallowing and pain and the physician-related
assessments demonstrated the importance of patient
reported outcomes and motivated their use in sub- 2 Vinorelbine and Gemcitabine
sequent trials (Radiation Therapy Oncology Group
RTOG 0617). In addition to paclitaxel, other third generation che-
Phase III evidence demonstrating the benefit of motherapy agents, such as vinorelbine and gemcita-
concurrent chemoradiation over sequential chemora- bine, have been incorporated in concurrent
diation for locally advanced NSCLC was initially chemoradiation regimens for locally advanced
provided by studies from the West Japan Lung Cancer NSCLC. Vinorelbine, a vinca alkaloid and potent
Group (WJLCG) and the Radiation Therapy Oncol- inhibitor of microtubule polymerization, gained
ogy Group (RTOG) (Furuse et al. 1999; Curran et al. attention because of its broad spectrum of antitumor
2003). However, both of these studies employed activity seen in preclinical studies. Vinorelbine was
second generation chemotherapy agents. In contrast to more active than other vinca alkaloids in almost all
second generation agents that could be delivered at models tested. Additionally, in a murine model
full systemic dose concurrently with radiation, third vinorelbine was shown to have synergy with cisplatin
generation chemotherapy agents had to be adminis- (Cros et al. 1989). Vinorelbine was the first third
tered at less than systemic doses during radiation. generation agent to receive FDA approval for
There was concern that third generation agents given NSCLC. Crawford et al. reported improved survival
at reduced doses during periods of irradiation were with vinorelbine compared to fluorouracil and leu-
not optimally treating micrometastatic disease. The covorin in patients with advanced NSCLC (Crawford
disappointing results reported in the CALGB 39801 et al. 1996). Studies also demonstrated that vinorel-
study may have added to the concern about using bine plus cisplatin was superior to either agent alone
third generation agents for chemoradiotherapy. To in the context of advanced disease (Le Chevalier
assess the benefit of replacing full dose second gen- et al. 1994; Wozniak et al. 1998). Additionally, Le
eration agents with more frequent radiosensitizing Chevalier et al. showed the superiority of cisplatin
dose third generation agents for concurrent chemo- and vinorelbine over the previous French standard
radiotherapy, the West Japan Thoracic Oncology of cisplatin and vindesine for advanced NSCLC
Group (WJTOG) conducted a phase III study (Le Chevalier et al. 1994). Considering another third
(Yamamoto et al. 2010). The study compared three generation chemotherapy agent that acts on microtu-
arms: (control arm) cisplatin, vindesine, and mito- bules, paclitaxel, was recently shown to radiosensitize
mycin with concurrent radiation followed by cis- cells, investigators studied vinorelbine’s interaction
platin, vindesine, and mitomycin consolidation; with radiation. In the human lung carcinoma cell
carboplatin and irinotecan with concurrent radiation line NCl-H460, vinorelbine was shown to potentiate
followed by carboplatin and irinotecan consolidation; radiation in a cell cycle dependent manner with the
298

Table 3 Summary of selected docetaxel chemoradiation studies


Reference Study Patients Induction Concurrent Consolidation MST Comments
(months)
Gandara et al. (2003) S9504, phase 83 – P/E/61 Gy Doce 26 Tolerable regimen with encouraging MST.
II
Vergnenègre et al. GFPC Phase 40 P/Vrb Doce/66 Gy – 13 Acceptable toxicity but modest survival rates.
(2005) II
Sakai et al. (2004) Phase II 32 – Bi-weekly Carbo/Doce Carbo/Doce 27 Concurrent bi-weekly CT regimen and RT was
60 Gy active with manageable toxicity.
Hanna et al. (2008) HOG, phase 203 – P/E/59.4 Gy – 23.2 Consolidation doce increased toxicity but not
III – P/E/59.4 Gy Doce 21.2 survival.

Kelly et al. (2008) S0023, phase 243 – P/E/61 Gy Doce 35 Gefinitib did not improve survival. Survival
III – P/E/61 Gy Doce/ 23 time compares favorably to other phase III
Gefinitib studies.

Jain et al. (2009) Phase II 61 – Carbo/Doce/63 Gy Carbo/Doce 12 Regimen has acceptable toxicity but
disappointing survival data.
Movsas et al. (2010) Phase II 83 – P/E/62 Gy Gem 16.1 Both arms were tolerable. In the Gem/Doce
– P/E/62 Gy Gem/Doce 29.5 arm, survival rate is higher than previous trials.

Segawa et al. (2010) Phase III 200 – M/V/P/60 Gy – 23.7 Trend toward improved survival in the
– P/Doce/60 Gy – 26.8 P/Doce arm.

Senan et al. (2011) Phase II 70 P/Doce P/Doce/66 Gy – 63.2% Both arms merit further testing in small volume
(1 year) (V20 \ 35%) disease.
– P/Doce/66 Gy P/Doce 65.5%
(1 year)
MST median survival time, P cisplatin, E etoposide, Carbo carboplatin, Doce docetaxel, SWOG Southwest Oncology Group, Hoosier Oncology Group
R. Bland et al.
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 299

greatest potentiation ocurring when the cells were in that gemcitabine might have the greatest radiation
the G2 phase (Edelstein et al. 1996). associated toxicity. Indeed, esophagitis was most
Gemcitabine, another third generation agent, is an pronounced in the gemcitabine arm with a 35% rate of
analog of the pyrimidine antimetabolite cytosine ara- grade 3 and a 17% rate of grade 4 toxicity. However,
binoside (ara-C). However, gemcitabine accumulates radiation pneumonitis was no more common in the
within tumor cells at much higher levels and for longer gemcitabine arm than the other two arms. The
periods of time than the active metabolite of ara-C investigators compared the overall median survival of
(Heinemann et al. 1988; Hertel et al. 1990). Like pac- 17 months seen in this trial to the median survival
litaxel and vinorelbine, gemcitabine was shown to be times from two other studies that used second gen-
modestly superior to the second generation chemo- eration agents for concurrent chemoradiation (Furuse
therapy agents when used for advanced NSCLC et al. 1999; Curran et al. 2003). This comparison led
(Ginsberg et al. 2001; Bunn and Kelly 1998; Hoffman the investigators to conclude that the survival in this
et al. 2000; Johnson 1999; Bonomi et al. 2000; study was greater than that seen in previous CALGB
Giaccone et al. 1998; Le Chevalier et al. 1994; Wozniak trials most likely due to the benefit provided by
et al. 1998; Sandler et al. 2000). Preclinical studies concurrent chemoradiation and less likely due to the
revealed that gemcitabine exhibits cytotoxic synergy newer chemotherapy agents.
with several agents including cisplatin (Nakamura et al. A group from the Czech Republic employed
1995; Tanzer et al. 1995). Additionally, laboratory vinorelbine in a study evaluating the benefit of con-
research showed gemcitabine to be a radiation sensi- current chemoradiation (Zatloukal et al. 2004). This
tizer of colon and pancreatic cancer cells (Shewach study randomized patients to four cycles of cisplatin
et al. 1994; Lawrence et al. 1996). and vinorelbine with radiotherapy commencing on the
For the same reasons as paclitaxel, vinorelbine and second cycle (concurrent arm) or after completion of
gemcitabine each combined with a platinum com- chemotherapy (sequential arm). Consistent with pre-
pound were attractive regimens to use for concurrent vious phase III studies, the median survival was sig-
chemoradiation for locally advanced NSCLC. These nificantly longer in the concurrent arm (16.6 months)
third generation regimens had improved activity compared to the sequential arm (12.9 months). The
over second generation regimens in advanced NSCLC concurrent arm was more toxic but the added toxicity
trials and preclinical research demonstrated they did not affect the delivery of therapy and there were
were radiation sensitizers. In this context, CALGB no treatment related deaths. These results added to the
designed a three arm randomized phase II trial growing body of evidence that concurrent chemora-
(CALGB 9431) to evaluate paclitaxel, vinorelbine, diotherapy yields improved survival over sequential
and gemcitabine in combination with cisplatin as chemoradiotherapy at the cost of increased, but
induction and concurrent chemoradiotherapy (Vokes manageable toxicity.
et al. 2002). This study and other select studies uti- A randomized trial by the French Pneumonology
lizing vinorelbine and/or gemcitabine are summarized Group (GFPC) provided evidence that vinorelbine
in Table 2. Patients in each arm of the CALGB 9431 was a better drug than vindesine to combine with
study received induction chemotherapy with cisplatin cisplatin and mitomycin for the treatment of advanced
and one of the newer agents followed by cisplatin and NSCLC (Perol et al. 1996). Subsequently, the Groupe
the same newer agent concurrently with 66 Gy of Lyon-Saint-Etienne d’Oncologie Thoracique (GLOT)
radiotherapy. Notably, this study was conducted and GFPC jointly designed a phase III trial using
before there was evidence discouraging the use of vinorelbine with cisplatin to compare sequential and
induction chemotherapy before concurrent chemora- concurrent chemoradiotherapy for locally advanced
diation (Belani et al. 2005; Vokes et al. 2007). This NSCLC (Fournel et al. 2005). The sequential arm
study revealed that all three regimens can be safely treated patients with cisplatin and vinorelbine induc-
delivered and have similar efficacy with median sur- tion chemotherapy followed by radiotherapy. The
vival times for the gemcitabine, paclitaxel, and concurrent arm treated patients with cisplatin and
vinorelbine containing arms of 18.3, 14.8, and etoposide and concurrent radiotherapy followed by
17.7 months, respectively. However, the three arms cisplatin and vinorelbine consolidation chemotherapy.
exhibited different toxicity profiles. It was suspected Although not statistically significant, the median
300

Table 4 Summary of selected pemetrexed chemoradiation studies


Reference Study Patients Induction Concurrent Consolidation MST Comments
(months)
Gadgeel et al. Phase II 21 – P/Pem/66 Gy Doce NR RR 53%
(2008)
Gauden et al. Phase II 25 – Carbo/Doce/ Pem 22 RR 76%
(2009) 60 Gy
Germonpre et al. Phase II 30 P/Pem Pem/60 Gy – NR RR 69%
(2009) – Pem/60 Gy P/Pem NR
Govindan et al. CALGB 30407, 99 – Carbo/Pem/70 Gy Pem 22 Both regimens of full dose
(2008) Phase II – Carbo/Pem/Cetux/ Pem 22 Carbo/Pem/RT are feasible
70 Gy and tolerable.

Brade et al. (2010) Phase II 39 – P/Pem/61-66 Gy P/Pem 20 Full dose P/Pem/RT is active
and tolerable.
Choy et al. (2010) Phase II 76 – Carbo/Pem/64- Pem NR RR 38-41%
68 Gy
– P/Pem/64-68 Gy Pem NR
Xu et al. (2010) Phase II 21 – Carbo/Pem/60- Carbo/Pem NR RR 86%
66 Gy
Vokes et al. PROCLAIM, 600, nonsquamous – P/Pem/66 Gy Pem NA Currently evaluating benefit
(2009) Phase III (planned) – P/E/66 Gy P/E or P/Vrb or NA of full dose pem in concurrent
Carbo/Pac CTRT.

MST median survival time, P cisplatin, Pem pemetrexed, E etoposide, Carbo carboplatin, Vrb vinorelbine, CALGB Cancer and Leukemia Group B, CTRT chemoradiation
R. Bland et al.
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 301

Table 5 Summary of selected studies with altered fractionation or escalated radiation doses
Reference Study Patients Induction Concurrent Consolidation MST Comments
(months)
Choy et al. Phase II 43 – Carbo/Pac Carbo/Pac 14.3 Regimen is well
(2000) 1.2 Gy BID/ tolerated with
69.6 Gy encouraging response
rate.
Reguart et al. Phase II 37 Plat/Gem Plat/Vrb/ Plat/Gem 15.4 Results are promising
(2004) 1.8 ? 0.88 Gy although toxicity is
boost/61.64 Gy considerable.
Casas et al. Phase II 32 Carbo/ Daily Pac/ Carbo/Pac 16.9 Acceptable efficacy and
(2011) Pac 1.8 ? 0.88 Gy less toxicity than the
boost/61.64 Gy trial in the row above.
Patel et al. Phase II 35 Carbo/ Topotecan/ – 17.9 Encouraging survival
(2007) Vrb 2 Gy bid, every with no severe
other week, esophagitis.
60 Gy
Schild et al. Phase I 15 – Carbo/Pac/70- – 37 MTD of RT was 74 Gy.
(2006) 78 Gy Phase II portion is
underway.
Stinchcombe Phase I/ 62 Carbo/ Carbo/Pac/60- – 25 Dose escalation
et al. (2008) II Pac 74 Gy compares favorably to
previous trials.
Socinski CALGB 69 Carbo/ Carbo/Pac/ – 24.3 Encouraging MST and
et al. (2008) 30105, Pac 74 Gy toxicity profile in arm 1.
Phase II Carbo/ Gem/74 Gy – 12.5 Arm 2 was too toxic.
Gem (closed
early)
Bradley et al. RTOG 53 – Carbo/Pac/ Optional 25.9 Among the best MSTs
(2010) 0117, 74 Gy in RTOG studies.
Phase II
MST median survival time, P cisplatin, Pem pemetrexed, E etoposide, Carbo carboplatin, Vrb vinorelbine, CALGB Cancer and
Leukemia Group B, CTRT chemoradiation

survival time for the concurrent arm was greater than two cycles of cisplatin and paclitaxel followed by
that seen in the sequential arm (16.3 versus cisplatin and vinorelbine with concurrent radiation
14.5 months). Despite 32% esophageal toxicity in the (induction/concurrent arm) or cisplatin and vinorel-
concurrent arm compared to 3% in the sequential arm, bine with concurrent radiation followed by two cycles
the results support the existing evidence for the use of of cisplatin and paclitaxel (concurrent/consolidation
concurrent over sequential chemoradiotherapy. arm). This trial observed similar toxicities and
Notably, since at the time of the design of this study response rates in both arms but greater median sur-
cisplatin and vinorelbine was not authorized in France vival time in the induction/concurrent arm (19.3
for concurrent use with radiation, this trial relied on a versus 16.9 months).
second generation regimen, cisplatin and etoposide, While induction and consolidation combinations of
for the chemotherapy given concurrently with radia- carboplatin and gemcitabine had been studied, there
tion. However, the GLOT subsequently designed a were no definitive studies establishing the use of
phase II study that did utilize cisplatin and vinorelbine gemcitabine and carboplatin given concurrently
given concurrently with radiation (Fournel et al. with radiotherapy for locally advanced NSCLC.
2006). The goal of this study was to evaluate the To determine the maximum tolerated dose of gem-
sequencing of chemotherapy and concurrent chemo- citabine for concurrent delivery with carboplatin and
radiotherapy. Patients were randomized to receive radiotherapy, Choy et al. conducted RTOG 0017
302 R. Bland et al.

(Choy et al. 2009). Considering the favorable to collateral normal tissue and allowing for dose
response rates of gemcitabine and paclitaxel as single escalation. The arm using gemcitabine was closed
agents in NSCLC (Gatzemeier et al. 1996; Abratt early secondary to treatment-related deaths. In two of
et al. 1994; Hatcher et al. 1998) as well as their the three patients that died, a review of the radiation
radiation enhancing properties, this phase I study plans showed greater than 40% of the total lung
simultaneously evaluated the maximum tolerated volume was receiving at least 20 Gy. It has been
dose of gemcitabine and paclitaxel for concurrent shown that V20 (volume of organ receiving greater
chemoradiation. This study was known as the than 20 Gy) is a useful parameter for predicting
‘‘ping-pong trial’’ as dose cohorts were accrued to the radiation pneumonitis (Graham et al. 1999). Given
two treatment sequences in an alternating manner. this experience, the investigators concluded that any
Patients in both sequences received carboplatin and future trial using aggressive radiotherapy should
gemcitabine consolidation chemotherapy. The study include prospective definitions of healthy tissue con-
found the maximum tolerated dose of gemcitabine to straints. Gemcitabine is known to be an effective
be 450 mg/m2/week when given concurrently with radiation sensitizer that may also excessively radio-
carboplatin (AUC 2) and thoracic radiation (63 Gy). sensitize normal tissue (Blackstock et al. 2001, 2006).
The seven patients receiving this regimen experienced The early closure of the gemcitabine containing arm
two dose limiting toxicities (grade 4 neutropenia and supports the notion that gemcitabine is associated
grade 3 esophagitis). The starting doses of gemcitabine with greater toxicity than other third generation
(300 mg/m2/week) and paclitaxel (30 mg/m2/week) agents when used concurrently with radiation. This
were too toxic to be administered concurrently with CALGB study is being followed up by a phase III trial
63 Gy of thoracic radiotherapy. The median survival comparing standard dose (60 Gy) versus high-dose
times of 13.3 months for the concurrent carboplatin (74 Gy) conformal radiotherapy with concurrent
and gemcitabine sequence and 12.9 months for the and consolidation carboplatin and paclitaxel (RTOG
gemcitabine and paclitaxel concurrent sequence were 0617).
lower than anticipated.
In the aforementioned CALGB 9431 trial that
evaluated paclitaxel, vinorelbine, and gemcitabine in 3 Docetaxel
combination with cisplatin as induction and concur-
rent chemoradiotherapy, the gemcitabine arm had Adding chemotherapy sequentially to radiation
similar activity as the other two arms and manageable therapy improved survival in locally advanced
toxicity (Vokes et al. 2002). After several studies NSCLC patients by reducing distant progression
suggested that 74 Gy could be safely and effectively (Sause et al. 1995). Compared to sequential, con-
delivered (via three-dimensional (3D) planning) after current chemoradiation has improved survival by
induction chemotherapy and with concurrent chemo- reducing local recurrence with no effect on distant
therapy (Blackstock et al. 2001, 2006; Rosenman control (Schaake-Koning et al. 1992). Since con-
et al. 2002; Socinski et al. 2000), the CALGB con- current phase chemotherapy is limited by toxicity,
ducted a multi-institutional phase II study (CALGB many trials have implemented induction or consol-
30105) to evaluate two similar regimens: (arm A) idation chemotherapy in hopes of reducing distant
induction carboplatin and paclitaxel followed by progression.
carboplatin and paclitaxel with concurrent radiother- Docetaxel, a taxane and third generation chemo-
apy (74 Gy) and (arm B) induction carboplatin and therapy agent, is an attractive agent for consolidation
gemcitabine followed gemcitabine and radiation chemotherapy for several reasons. Preclinical models
therapy (74 Gy) (Socinski et al. 2008). Arm A had an suggested docetaxel has molecular mechanisms of
impressive median survival time of 24.3 months and action, such as p27 induction and Bcl-2 phosphoryla-
1-year survival rate of 66.7% which the investigators tion, which are thought to be most effective after cells
attributed to using 3D planning to deliver a higher have been exposed to DNA-damaging chemoradiation.
dose of radiation compared to previous trials. 3D Additionally, these models showed that docetaxel
planning allows for increased conformality of dose has activity against tumors with a p53 mutation
around tumor with margin, potentially reducing dose (Blagosklonny et al. 1996; Gumerlock et al. 1999).
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 303

Out of the newer agents, docetaxel demonstrated the Using H460 human lung carcinoma cells, Amorino
greatest single agent activity in chemotherapy-naïve et al. found that combination docetaxel and carbo-
patients with advanced NSCLC (Fossella 1999). platin enhances radiation’s effect on colony forming
Additionally, docetaxel has been shown in phase II units more than either drug separately (Amorino et al.
studies to have second-line activity in patients who 1999). Remarkably, the redistribution of cells into the
failed prior cisplatin-based treatment regimens for radiosensitive G2/M phase observed after paclitaxel
NSCLC (Fosella et al. 2000). Based on these preclinical exposure was not seen after docetaxel exposure.
studies and encouraging clinical results, the SWOG Considering these encouraging preclinical findings,
evaluated docetaxel for consolidation chemotherapy. Choy et al. performed a phase I study of weekly
In SWOG 9504, a phase II study, patients were treated docetaxel and carboplatin with concurrent radiother-
with full dose cisplatin and etoposide with con- apy in patients with locally advanced NSCLC (Choy
current radiation followed by consolidation docetaxel et al. 2001). The study showed the dose limiting
(Gandara et al. 2003). The important points of the toxicity was esophagitis and the maximum tolerated
SWOG 9504 study and other select docetaxel studies dose of docetaxel was 20 mg/m2 per week with
are highlighted in Table 3. weekly carboplatin (AUC 2) and concurrent radiation.
SWOG 9019, the predecessor trial to SWOG 9504, Subsequently, a phase II study was performed to
evaluated cisplatin and etoposide with concurrent assess the safety and efficacy of weekly docetaxel
radiation followed by consolidation cisplatin and (20 mg/m2) and carboplatin (AUC 2) administered
etoposide (Albain et al. 2002). The SWOG 9504 trial concurrently with radiotherapy (63 Gy) and followed
used the same eligibility criteria, staging, documen- by docetaxel and carboplatin consolidation (Jain et al.
tation procedures for T4 or N, and concurrent che- 2009). While the toxicity was acceptable, the survival
moradiation regimen as was used in SWOG 9019. rates of 45% at 1 year and 20% at 2 years were
SWOG 9504 replaced the consolidation cisplatin and disappointing.
etoposide regimen used in SWOG 9019 with doce-
taxel with the idea that a comparison of the two trials
could provide some relative information on the effi- 4 Irinotecan
cacy and safety of docetaxel as a consolidative regi-
men. The treatment in SWOG 9504 was found to be Camptothecin and its derivatives, such as irinotecan,
generally tolerable and resulted in an impressive target the DNA relaxing enzyme topoisomerase I
median survival of 26 months compared to 15 months (Hsiang et al. 1985; Hsiang and Liu 1988; Andoh
seen in SWOG 9019. et al. 1987; Nitiss and Wang 1988). Camptothecin
The Hoosier Oncology Group (HOG) took the next stabilizes the topoisomerase I-DNA intermediate that
step in evaluating consolidation chemotherapy with is formed when topoisomerase I cleaves DNA to
docetaxel by designing a prospective phase III study. allow for uncoiling. The stabilized intermediate
This study randomized patients to either (1) cisplatin interacts with the progressing replication fork and
and etoposide with concurrent radiation or (2) cis- results in a DNA double-strand break that is fatal to
platin and etoposide with concurrent radiation fol- the cell. Several studies have shown camptothecin
lowed by consolidation docetaxel. The median sensitizes cells to radiation in vitro and in vivo
survival time for the docetaxel arm was 21.5 months (Kudoh et al. 1993; Rich and Kirichenko 1998).
and was not statistically significantly different from Omura et al. found enhanced cell kill with combina-
the median survival time of 24.1 months seen in the tion radiation and irinotecan with the largest gains
arm without docetaxel consolidation. Additionally, occurring when irinotecan was administered just
the patients that received consolidation docetaxel before or just after irradiation (Omura et al. 1997).
experienced greater toxicity including an increased Another study showed that comptothecin derivatives
rate of hospitalization and premature death. The radiosensitized MCF-7 breast cancer cells in a dose-
investigators concluded that consolidation docetaxel dependent manner with the greatest effect seen when
does not improve survival but does add toxicity and cells were exposed to the drug before or during
therefore should no longer be used for patients with radiation (Chen et al. 1997). Additionally, in a study
unresectable stage III NSCLC (Hanna et al. 2008). using murine fibrosarcomas, radiation enhancing
304 R. Bland et al.

effects were seen when topotecan, another campto- attenuated dose. Pemetrexed, unlike other third gen-
thecin derivative, was administered 2 and 4 h before eration chemotherapy agents, is capable of being
radiation but not when administered 2 h after radia- administered at full systemic dose concurrently with
tion. The consistent findings in these preclinical thoracic radiotherapy. Consequently, investigators
studies suggested irinotecan be administered before or have hypothesized that pemetrexed could be helpful
during radiotherapy take full advantage of its radio- in reducing distant progression in patients treated with
sensitizing properties. concurrent chemoradiation.
After studies demonstrated concurrent chemoradi- Pemetrexed, a new generation antifolate, acts by
ation with irinotecan was safe and active in locally inhibiting several enzymes involved in nucleotide
advanced NSCLC (Takeda et al. 1999; Saka et al. synthesis, particularly thymidilate synthase (Joerger
1997), several phase I trials evaluated the addition of et al. 2010; Solomon and Bunn 2005). This agent has
carboplatin in the concurrent phase Uejima 2002 been shown to have activity in multiple tumor types
(Uejima 2002; Yamada et al. 2002; Chakravarthy including NSCLC (Clarke et al. 1997; Rusthoven
et al. 2000). These preliminary studies reported nau- et al. 1999; Smit et al. 2003). Pemetrexed appears to
sea, vomiting, and esophagitis as the dose limiting be a good candidate for concurrent chemoradiation as
toxicities of weekly irinotecan and carboplatin with studies have suggested a synergy between premetr-
concurrent radiation. Additionally, these studies pro- exed and radiotherapy in vitro (Bischof et al. 2002).
vided initial evidence of clinical activity for this In a phase I study, Seiwert et al. demonstrated that
regimen, with response rates of 60-70%. Concurrent full dose pemetrexed (500 mg/m2) and carboplatin
chemoradiation with irinotecan has been studied in (AUC 5 or 6) with concurrent thoracic radiation was
the phase III setting in the aforementioned WJTOG tolerable and active in patients with locally advanced
trial that compared second and third generation che- NSCLC or esophageal cancer (Seiwert et al. 2007).
motherapy in the treatment of locally advanced Subsequently, several more phase I studies have
NSCLC (Yamamoto et al. 2010). Arm B of this study provided additional evidence to support the safety of
utilized carboplatin and irinotecan for concurrent concurrent radiation with pemetrexed (Seiwert et al.
chemoradiation and consolidation chemotherapy. 2007; Bischof et al. 2010), cisplatin and pemetrexed
This arm’s median survival time (19.8 months) was (Brade 2010; Cardenal et al. 2009; Hensing et al.
not significantly different from the median survival 2009, 2010), or carboplatin and pemetrexed (Seiwert
times of the other two arms. However, the other et al. 2007; Machtay et al. 2008; Heinzerling et al.
experimental arm, which employed carboplatin and 2010). All of these phase I trials have achieved full-
paclitaxel for concurrent and consolidation chemo- dose concurrent phase pemetrexed with no trial
therapy, had a median survival time of 22 months and requiring early termination due to toxicity. These
the most favorable toxicity profile of the three arms. phase I studies exhibited acceptable rates of hema-
The WJTOG concluded this arm should be a standard tologic toxicity, radiation pneumonitis, and esopha-
regimen in the treatment of locally advanced NSCLC. gitis leading to additional studies in the phase II
setting. Phase II studies testing pemetrexed-platin
combinations given concurrently with radiation for
5 Pemetrexed locally advanced NSCLC resulted in promising
median survival times of 20–22 months (Brade 2010;
A meta-analysis of randomized trials showed that Govindan et al. 2008). Three randomized trials have
concurrent chemoradiation improved overall survival reported the consistent finding of a favorable treat-
and locoregional progression when compared to ment by histology interaction for pemetrexed in
sequential chemoradiation (Auperin et al. 2007). patients with advanced lung cancer of nonsquamous
However, in this analysis the 3-year rate of distant histology (Peterson et al. 2007; Ciuleanu et al. 2008;
progression was similar for concurrent and sequential Scagliotti et al. 2008). Considering this histologic bias
chemoradiation at 39.5 and 39.1%, respectively. In and the promising results from phase II studies of
the trials studied in this meta-analysis, concurrent concurrent chemoradiation with pemetrexed-platin
phase chemotherapy was either a second generation regimens, investigators designed the PROCLAIM
agent at full dose or a third generation agent at an study. This currently ongoing phase III study is
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 305

evaluating pemetrexed and cisplatin with concurrent compare standard dose (60 Gy) versus high-dose
radiotherapy followed by consolidation pemetrexed (74 Gy) conformal radiotherapy with concurrent and
for NSCLC of other than predominately squamous consolidation carboplatin and paclitaxel in patients
cell histology (Vokes et al. 2009). A summary of with stage IIIA/IIIB NSCLC (RTOG 0617). Table 5
select pemetrexed trials is provided in Table 4. highlights the important aspects of select dose esca-
lation trials as well as trials that have examined novel
radiation dose fractioning.
6 Third Generation Agents and Dose In summary, our chapter has described some of the
Escalation rationale for combining systemic cytotoxic agents with
radiation in the treatment of lung malignancies in the
Most of the recent studies of concurrent chemoradi- definitive setting. We have offered a view of the
ation for locally advanced NSCLC have explored mechanisms by which systemic agents have synergized
novel chemotherapy agents and additional chemo- with radiation to promote improved tumor outcomes.
therapy outside the concurrent phase. With 2D radi- A number of systemic agents have also been described
ation planning techniques, dose escalation to improve that were ultimately too toxic or had inferior efficacy
local tumor control has been impeded by inaccurate when given concurrently with radiation. From the vast
tumor targeting and the associated injury to healthy number of studies presented, it is obvious that con-
tissue. Consequently, 60 Gy has remained the stan- current treatment is the optimal one with regards to the
dard dose since it was established in RTOG 73-01 greatest survival benefit in stage III lung cancer patients
(Perez et al. 1982, 1987). However, with the devel- who cannot undergo resection. However, it is also
opment of 3D planning techniques, the tumor volume obvious that much more can be done for this group of
can be more accurately targeted and dose-volume patients. Current interest has been generated in esca-
calculations can be used to limit toxicity to healthy lating radiation dose with concurrent systemics in the
tissue (Lee et al. 2006). Using 3D planning, the setting of improved radiation delivery methods, limit-
RTOG, North Central Cancer Treatment Group ing toxicity to surrounding normal tissue. Another
(NCCTG), and CALGB have explored delivering paradigm taking shape is to combine targeted agents
escalated doses of thoracic radiation concurrently with cytotoxic chemotherapy given concurrently with
with weekly carboplatin and paclitaxel. The phase I radiation to inactivate multiple oncogenic pathways or
component of RTOG 0117 determined that 74 Gy reduce the effectiveness of those biochemical pathways
was the maximum tolerated dose with concurrent effectuating resistance. Finally, there has been some
weekly carboplatin and paclitaxel (Bradley et al. thought to combining systemic agents with stereotactic
2010). In the phase II component, patients receiving radiation for early stage lung lesions in the hopes of
weekly carboplatin and paclitaxel with concurrent improving disease free survival. In general, there is a
radiation to 74 Gy had a median survival time of great need for continuing to optimize concurrent ther-
25.9 months. Of note, out of 53 patients 12 had grade apy from both the chemotherapy side as well as the
3 or greater lung toxicity. A prospective phase I study radiation side in promoting improved distant and local
by the NCCTG reported results for 13 patients treated control, respectively.
to 70, 74, or 78 Gy (Schild et al. 2006). In this study,
the lower two doses were well tolerated while 78 Gy
was not. The median survival time for these 13
References
patients was 37 months. Patients in arm A of the
previously discussed CALGB 30105 study received
Abratt RP, Bezwoda WR, Falkson G et al (1994) Efficacy and
induction carboplatin and paclitaxel followed by safety profile of gemcitabine in non-small-cell lung cancer:
concurrent carboplatin and paclitaxel with 74 Gy a phase II study. J Clin Oncol 12:1535–1540
radiotherapy (Socinski et al. 2008). This treatment Akerley W, Herndon JE Jr, Lyss AP, Choy H, Turrisi A,
regimen resulted in a median survival time of Graziano S, Williams T, Zhang C, Vokes EE, Green MR
(2005) Induction paclitaxel/carboplatin followed by con-
24.3 months and manageable toxicities. Based on current chemoradiation therapy for unresectable stage III
these preliminary studies, the NCCTG, CALGB, and non-small-cell lung cancer: a limited-access study–CALGB
ROTG have jointly initiated a phase III trial to 9534. Clin Lung Cancer 7(1):47–53
306 R. Bland et al.

Albain KS, Crowley JJ, Turrisi AT et al (2002) Concurrent Blanco R, Solé J, Montesinos J, Mesía C, Algara M, Terrassa J,
cisplatin, etoposide plus radiotherapy for pathologic stage Gay M, Domenech M, Bastus R, Bover I, Nogué M, Vadell
IIIB non-small cell lung cancer: A Southwest Oncology C, ACROSS (2008) Induction chemotherapy with cisplatin
Group phase II study (S9019). J Clin Oncol 20:3454–3460 and gemcitabine followed by concurrent chemoradiation
Amorino GP, Hamilton VM, Choy H (1999) Enhancement of with twice-weekly gemcitabine in unresectable stage III
radiation effects by combined docetaxel and carboplatin non-small cell lung cancer: final results of a phase II study.
treatment in vitro. Radiat Oncol Investig 7(6):343–352 Lung Cancer 62(1):62–71
Andoh T, Ishii K, Suzuki Y et al (1987) Characterization of a Bonomi P, Kim K, Fairclough D et al (2000) Comparison of
mammalian mutant with a camptothecin-resistant DNA survival and quality of life in advanced non-small-cell lung
topoisomerase I. Proc Natl Acad Sci USA 84:5565–5569 cancer patients treated with two dose levels of paclitaxel
Auperin A, Rolland E, Curran WJ et al., on behalf on the combined with cisplatin versus etoposide with cisplatin:
NSCLC Collaborative Group (2007) Concomitant radio- results of an Eastern Cooperative Oncology Group trial.
chemotherapy (RT-CT) versus sequential RT-CT in J Clin Oncol 18:623–631
locally advanced non-small cell lung cancer (NSCLC). A Brade AM (2010) A phase II study of concurrent pemetrexed/
meta-analysis using individual patient data from random- cisplatin/radiation (RT) for unresectable stage IIIA/b non
ised clinical trials. J Thorac Oncol 2(8 suppl 4):S310 small cell lung cancer (NSCLC) [abstract 7087]. J Clin
Begg AC, VaderKolk PJ, Edmondt J et al (1987) Radiosensiti- Oncol 28:s15
zation in vitro by cis-diammine (1, 1 -cyclobutanedicarboxy- Brade A, Bezjak A, MacRae R, Laurie S, Sun A, Cho J, Leighl
alto) platinum(II) (carboplatin, JM8) and etylnediammine N, Pearson S, Southwood B, Wang L, McGill S, Iscoe N,
malnatoplatinum(II) (JM40). Radiat Oncol 9:157–165 Shepherd FA (2011) Phase I trial of radiation with
Begg AC, Stewart FA, Dewitt L et al (1989) Interaction concurrent and consolidation pemetrexed and cisplatin in
between cisplatin and radiation in experimental rodent patients with unresectable stage IIIA/B non-small-cell lung
tumors and normal tissues. In: Hill B, Bellamy A (eds) cancer. Int J Radiat Oncol Biol Phys 79(5):1395–1401
Antitumor Drug Radiation Interactions. CRC Press, Boca Bradley J, Bae K, Graham M et al (2010) Primary analysis of
Raton, FL, pp 154–510 the phase II component of a phase I/II dose intensification
Belani CP (1993) Multimodality management of regionally study using three-dimensional conformal radiation therapy
advanced non-small-cell lung cancer. Semin Oncol and concurrent chemotherapy for patients with inoperable
20:302–314 non-small cell lung cancer: RTOG 0117. J Clin Oncol
Belani CP, Aisner J, Day R et al (1997) Weekly paclitaxel 28:2475–2480
and carboplatin with simultaneous thoracic radiotherapy Bunn PA Jr, Kelly K (1995) A phase I study of carboplatin
for locally advanced non-small cell lung cancer: Three and paclitaxel in non-small-cell lung cancer: a university
year follow-up. Proc Am Soc Clin Oncol 16:448a (abstr of colorado cancer center study. Semin Oncol 22(suppl 9):
1608) 2–6
Belani CP, Choy H, Bonomi P et al (2005) Combined Bunn PA Jr, Kelly K (1998) New chemotherapeutic agents
chemoradiotherapy regimens of paclitaxel and carboplatin prolong survival and improve quality of life in non-small
for locally advanced non-small-cell lung cancer: a random- cell lung cancer: a review of the literature and future
ized phase II locally advanced multimodality protocol. directions. Clin Cancer Res 4:1087–1100
J Clin Oncol 23:5883–5891 Calabro-Jones PM, Fahey RC, Smoluk GD et al (1985)
Bischof M, Weber KJ, Blatter J, Wannenmacher M, Latz D Alkaline phosphatase promotes radioprotection and accu-
(2002) Interaction of pemetrexed disodium (ALIMTA, mulation of WR-1065 in V79–171 cells incubated in
multitargeted antifolate) and irradiation in vitro. Int J medium containing WR-27821. Int J Radiat Oncol Biol
Radiat Oncol Biol Phys 52:1381–1388 Phys 47:23–27
Bischof M, Herfarth K, Thomas M et al (2010) Phase 1/2 study Cardenal F, Arnaiz M, Moran T et al (2009) Concurrent
of pemetrexed in combination with carboplatin and con- chemoradiation with pemetrexed and cisplatin followed by
comitant thoracic radiotherapy in chemonai‹ ve patients with consolidation pemetrexed for patients with unresectable
inoperable Stage I-IIIb non-small cell lung cancer: a single- stage III NSCLC: preliminary results of a phase I study.
arm, open-label, monocentric phase 1/2 study. Deutsche IASLC 13th World Conference on Lung Cancer:P3.0007
Gesellschaft fur Hamatologie und Onkologie (abstract) (abstract)
Blackstock AW, Lesser GJ, Fletcher-Steede J et al (2001) Phase Casas F, Viñolas N, Ferrer F, Agustí C, Sanchez M, Maria
I study of twice-weekly gemcitabine and concurrent Gimferrer J, Lomeña F, Campayo M, Jeremic B (2011)
thoracic radiation for patients with locally advanced non- Long-term results of a phase II trial of induction paclitaxel-
small-cell lung cancer. Int J Radiat Oncol Biol Phys carboplatin followed by concurrent radiation therapy and
51:1281–1289 weekly paclitaxel and consolidation paclitaxel-carboplatin
Blackstock AW, Ho C, Butler J et al (2006) Phase Ia/Ib chemo- in stage III non-small cell lung cancer. J Thorac Oncol
radiation trial of gemcitabine and dose-escalated thoracic 6(1):79–85
radiation in patients with stage III A/B non-small cell lung Chakravarthy A, Choy H, Devore RF III et al (2000) Phase I
cancer. J Thorac Oncol 1:434–440 trial of outpatient fweekly irinotecan (CPT-11) and carbo-
Blagosklonny M, Schulte TW, Nguyen P et al (1996) Taxol- platin with concurrent radiation therapy for stage III
induced apoptosis and phosphorylation of Bcl-2 protein unresectable non-small cell lung cancer: a Vanderbilt
involves c-Raf-1 and represents a novel c-Raf-1 signal Cancer Center affiliate network (VCCAN) trial. Proc Am
transduction pathway. Cancer Res 56:1851–1854 Soc Clin Oncol 18:1–2040
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 307

Chen AY, Okunieff P, Pommier Y et al (1997) Mammalian advanced non-small cell lung cancer (NSCLC). Proc Am
DNA topoisomerase I mediates the enhancement of radia- Soc Clin Oncol 16:465a (abstr 1670)
tion cytotoxicity by camptothecin derivatives. Cancer Res Coughlin CT, Richmond RC (1989) Biologic and clinical
57:1529–1536 developments of cisplatin combined with radiation: Con-
Choy H Schwartzberg L, Dakhil S et al (2010) Ongoing phase cepts, utility, projections of new trials, and the emergence of
II study of pemetrexed plus carboplatin or cisplatin with carboplatin. Semin Oncol 16(suppl 6):31–43
concurrent radiation therapy followed by pemetrexed con- Crawford J, O’Rourke M, Schiller JH, Spiridonidis CH,
solidation in patients with favorable-prognosis inoperable Yanovich S, Ozer H, Langleben A, Hutchins L, Koletsky
stage IIIA/B non-small-cell lung cancer: interim update. A, Clamon G, Burman S, White R, Hohneker J (1996)
J Clin Oncol 2010 ASCO Annual Meeting Procededings 28 Randomized trial of vinorelbine compared with fluorouracil
(suppl 15):7082(abstract) plus leucovorin in patients with stage IV non-small cell lung
Choy H, Devore RF 3rd, Hande KR, Porter LL, Rosenblatt P, cancer. J Clin Oncol 14:2777–2784
Yunus F, Schlabach L, Smith C, Shyr Y, Johnson DH Cros S, Wright M, Roussakis C et al (1989) Experimental
(2000) A phase II study of paclitaxel, carboplatin, and antitumor activity of navelbine (5’-nor-anhydrovinblastine,
hyperfractionated radiation therapy for locally advanced vinorelbine). Semin Oncol 16(Suppl 4):15–20
inoperable non-small-cell lung cancer (a Vanderbilt Cancer Curran WJ, Scott CB, Langer CJ et al (2003) Longterm benefit
Center Affiliate Network Study). Int J Radiat Oncol Biol is observed in a phase III comparison of sequential versus
Phys 47(4):931–937 concurrent chemo-radiation for patients with unresected
Choy H, Jain AK, Moughan J, Curran W, Whipple G, Demas stage III NSCLC: RTOG 9410. Proc Am Soc Clin Oncol
WF, Ettinger DS (2009) RTOG 0017: a phase I trial of 22:621 (suppl; abstr 2499)
concurrent gemcitabine/carboplatin or gemcitabine/paclit- Davies AM, Chansky K, Lau DH, Leigh BR, Gaspar LE, Weiss
axel and radiation therapy (‘‘ping-pong trial’’) followed by GR, Wozniak AJ, Crowley JJ, Gandara DR, SWOG S9712
adjuvant chemotherapy for patients with favorable progno- (2006) Phase II study of consolidation paclitaxel after
sis inoperable stage IIIA/B non-small cell lung cancer. concurrent chemoradiation in poor-risk stage III non-small-
J Thorac Oncol 4(1):80–86 cell lung cancer: SWOG S9712. J Clin Oncol.
Choy H, Safran H (1995) Preliminary analysis of a phase II 24(33):5242–5246
study of weekly paclitaxel and concurrent radiation therapy Dillman RO, Seagren SL, Propert KJ et al (1990) A randomized
for locally advanced non-small-cell lung cancer. Semin trial of induction chemotherapy plus high-dose radiation
Oncol 22(suppl 9):55–57 versus radiation alone in stage III non-small-cell lung
Choy H, Rodriguez FF, Koester S et al (1993) Investigation of cancer. N Engl J Med 323:940–945
Taxol as a potential radiation sensitizer. Cancer Dillman RO, Herndon J, Seagren SL et al (1996) Improved
71:3774–3778 survival in stage III non-small-cell lung cancer: seven-year
Choy H, Akerley W, Safran H et al (1994) Phase I trial of follow-up of Cancer and Leukemia Group B (CALGB)
outpatient weekly paclitaxel and concurrent radiation ther- S433 trial. J Natl Cancer Inst 88:1210–1215
apy for advanced non-small-cell lung cancer. J Clin Oncol Divers SG, Spencer SA, Carey D, Busby EM, Hyatt MD,
12(12):2682–2686 Robert F (2005) Phase I/IIa study of cisplatin and gemcit-
Choy H, Akerley W, Safran H et al (1998) Multi-institutional abine as induction chemotherapy followed by concurrent
phase II trial of paclitaxel, carboplatin and concurrent chemoradiotherapy with gemcitabine and paclitaxel for
radiation therapy for locally advanced inoperable non-small locally advanced non-small-cell lung cancer. J Clin Oncol
cell lung cancer. J Clin Oncol 16:3316–3322 23(27):6664–6673
Choy H, DeVore RF, Hande KR, Porter LL, Rosenblatt PA, Douple EB (1993) Platinum chemotherapy for cancer treat-
Slovis B, LaPorte K, Shyr Y, Johnson DH (2001) Phase I ment. In: Brooks RR (ed) Noble Metals and Biological
trial of outpatient weekly docetaxel, carboplatin, concurrent Systems. CRC Press, Boca Raton, FL, pp 349–376
thoracic radiation therapy for stage III unresectable non- Edelstein MP, Wolfe LA III (1996) Duch DS: potentiation of
small cell lung cancer. Lung Cancer 34(3):441–449 radiation therapy by vinorelbine (Navelbine) in non-small
Choy H, Jain AK, Moughan J, Curran W, Whipple G, cell lung cancer. Semin Oncol 23:41–47
Demas WF, Ettinger DS (2009) RTOG 0017: a phase I Fosella F, DeVore R, Kerr RN et al (2000) Randomized phase
trial of concurrent gemcitabine/carboplatin or gemcitabine/ III trial of docetaxel versus vinorelbine or ifosfamide in
paclitaxel and radiation therapy (‘‘Ping-Pong Trial’’) fol- patients with advanced non small cell lung cancer previ-
lowed by adjuvant chemotherapy for patients with favorable ously treated with platinum containing chemotherapy reg-
prognosis inoperable stage IIIA/B non-small cell lung imens. J Clin Oncol 18:2354–2362
cancer. J Thoracic Oncol 4(1):80–86 Fossella FV (1999) Docetaxel in the treatment of non-small cell
Ciuleanu TE, Brodowicz T, Belani CP et al (2008) Mainte- lung cancer: review of single-agent trials. Semin Oncol
nance pemetrexed plus best supportive care (BSC) versus 26(suppl 16):17–23
placebo plus BSC: a phase III study. J Clin Oncol 26(suppl Fournel P, Robinet G, Thomas P et al (2005) Randomized
15):426s (Abstract 8011) phase III trial of sequential chemoradiotherapy compared
Clark JW, Santos-Moore AS, Choy H (1998) Sequencing of with concurrent chemoradiotherapy in locally advanced
taxol and carboplatinum therapy. Submitted 1995 AACR non-small-cell lung cancer: Groupe Lyon-Saint-Etienne
Annual Meeting 36:298 d’Oncologie Thoracique-Groupe Francais de Pneumo-
Clarke S, Boyer M, Millward M et al (1997) Phase II study of Cancerologie NPC 95–01 Study. J Clin Oncol 23:
LY231514, a multi-targeted antifolate, in patients with 5910–5917
308 R. Bland et al.

Fournel P, Vergnene0 gre A, Robinet G et al (2006) Induction or Hanna N, Neubauer M, Yiannoutsos C et al (2008) Phase III
consolidation chemotherapy with cisplatin (C) And paclit- trial of cisplatin, etoposide, and concurrent chest radiation
axel (P) plus concurrent chemo-radiation with cisplatin and with or without consolidation docetaxel in patients with
vinorelbine (V) for unresectable non-small cell lung cancer inoperable stage III nonsmall- cell lung cancer: The Hoosier
patients: randomized phase II trial GFPC-GLOT-IFCT 02- Oncology Group and U.S. Oncology. J Clin Oncol 26:
01. J Clin Oncol, ASCO Annual Meeting Proceedings Part I 5755–5760
24: 18S (June 20 Suppl), 7048, 2006 Hatcher N, Ranson M, Anderson H et al (1998) Phase III study
Furuse K, Fukuoka M, Kawahara M et al (1999) Phase III study of paclitaxel (Taxol) (T) versus best supportive care (BSC)
of concurrent versus sequential thoracic radiotherapy in in inoperable non-small cell lung cancer (NSCLC).
combination with mitomycin, vindesine, and cisplatin in Presented at the 23rd Congress of the European Society of
unresectable stage III non-small cell lung cancer. J Clin Medical Oncology, Athens, Greece
Oncol 17:2692–2699 Heinemann V, Hertel LW, Grindey GB et al (1988) Compar-
Gadgeel S, Ruckdeschel J, Wozniak A et al (2008) Pemetrexed ison of the cellular pharmacokinetics and toxicity of 2’, 2’-
and cisplatin with concurrent thoracic radiation therapy difluorodeoxycytidine and 1-beta-D-arabinofuranosylcyto-
(TRT) followed by docetaxel in stage III non-small cell lung sine. Cancer Res 48:4024–4031
cancer (NSCLC) patients (pts). In: ASCO Annual Meeting Heinzerling J, Choy H, Hughes R et al (2010) Toxicity and
Proceedings. J Clin Oncol 26 (abstract 7569) response of pemetrexed plus carboplatin or cisplatin with
Gandara DR, Chansky K, Albain KS et al (2003) Consolidation concurrent chest radiation therapy for patients with locally
docetaxel after concurrent chemoradiotherapy in stage IIIB advanced non-small cell lung cancer: a phase I trial.
non-small-cell lung cancer: phase II Southwest Oncology J Thorac Oncol 5:1391–1396
Group Study S9504. J Clin Oncol 21:2004–2010 Hensing T, Patel J, Marymont M et al. (2009) A phase I trial of
Gatzemeier U, Shepherd FA, Le Chevalier T et al (1996) cisplatin, pemetrexed, and radiation in patients with stage
Activity of gemcitabine in patients with non-small cell lung III non-small-cell lung cancer. IASLC 13th World Confer-
cancer: a multicentre, extended phase II study. Eur J Cancer ence on Lung Cancer:P3.019 (abstract)
32:243–248a Hertel LW, Boder GB, Kroin JS et al (1990) Evaluation of the
Gauden S, Haug G, Markos J et al (2009) Phase II study of antitumor activity of gemcitabine (2’, 2’-difluoro-2’- deox-
combined modality therapy with concurrent docetaxel and ycytidine). Cancer Res 50:4417–4422
carboplatin plus radiation therapy followed by consolidation Hirsh V, Soulieres D, Duclos M, Faria S, Del Vecchio P, Ofiara L,
pemetrexed chemotherapy for unresectable stage III non- Ayoub JP, Charpentier D, Gruber J, Portelance L, Souhami L
small-cell-lung cancer. IASCL 13th World Conference on (2007) Phase II multicenter trial with carboplatin and
Lung Cancer: P3.014 (abstract) gemcitabine induction chemotherapy followed by radiother-
Germonpre P, Janssens A, Galdermans D et al (2009) A apy concomitantly with low-dose paclitaxel and gemcitabine
randomized muticenter phase II study of concurrent for stage IIIA and IIIB non-small cell lung cancer. J Thorac
pemetrexed and radiotherapy with induction or adjuvant Oncol 2(10):927–932
cisplatin/pemetrexed in patients with unresectable stage III Hoffman PC, Mauer AM, Vokes EE (2000) Lung cancer.
non-small cell lung cancer (NSCLC) IASLC 13th World Lancet 355:479–485 (published erratum appears in Lancet
Conference on Lung Cancer: P3.015 (abstract) 355:1280, 2000)
Giaccone G, Splinter TA, Debruyne C et al (1998) Randomized Hsiang YH, Liu LF (1988) Identification of mammalian DNA
study of paclitaxel-cisplatin versus cisplatin-teniposide in topoisomerase I as an intracellular target of the anticancer
patients with advanced non-small-cell lung cancer: The drug camptothecin. Cancer Res 48:1722–1726
European Organization for Research and Treatment of Cancer Hsiang YH, Hertzberg R, Hecht S et al (1985) Camptothecin
Lung Cancer Cooperative Group. J Clin Oncol 16:2133–2141 induces protein-linked DNA breaks via mammalian DNA
Ginsberg R, Vokes EE, Rosenzweig K (2001) Non-small cell topoisomerase I. J Biol Chem 260:14873–14878
lung cancer. In: DeVita JV, Hellman S, Rosenberg SA (eds) Huber RM, Flentje M, Schmidt M, Pöllinger B, Gosse H,
Cancer-Principles, Practice of Oncology, 6th edn. JB Willner J, Ulm K, Group Bronchial Carcinoma Therapy
Lippinoctt Co, Philadelphia, PA, pp 925–982 (2006) Simultaneous chemoradiotherapy compared with
Govindan R, Bogart J, Wang X et al (2008) A phase II study of radiotherapy alone after induction chemotherapy in inoper-
pemetrexed, carboplatin and thoracic radiation with or able stage IIIA or IIIB non-small-cell lung cancer: study
without cetuximab in patients with locally advanced CTRT99/97 by the Bronchial Carcinoma Therapy Group.
unresectable non-small cell lung cancer: CALGB 30407– J Clin Oncol 24(27):4397–4404
Early evaluation of feasibility and toxicity. J Clin Oncol Jain AK, Hughes RS, Sandler AB, Dowlati A, Schwartzberg
26(18 suppl):401s (Abstract 7518) LS, Dobbs T, Schlabach L, Wu J, Muldowney NJ, Choy H
Graham MV, Purdy JA, Emami B et al (1999) Clinical dose- (2009) A phase II study of concurrent chemoradiation with
volume histogram analysis for pneumonitis after 3D treat- weekly docetaxel, carboplatin, and radiation therapy fol-
ment for non-small cell lung cancer (NSCLC). Int J Radiat lowed by consolidation chemotherapy with docetaxel and
Oncol Biol Phys 45:323–329 carboplatin for locally advanced inoperable non-small cell
Gumerlock PH, Mack PC, Manorek GH et al (1999) Bcl-2 lung cancer (NSCLC). J Thoracic Oncol 4(6):722–727
phosphorylation, p27 induction as potential p53-indepen- Joerger M, Omlin A, Cerny T, Früh M (2010) The role of
dent mechanisms of apoptotic response to taxanes in non- pemetrexed in advanced non small-cell lung cancer: special
small cell lung carcinoma. Proc Am Assoc Cancer Res focus on pharmacology and mechanism of action. Curr
40:739 (abstr 4879) Drug Targets 11:37–47
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 309

Johnson DH (1999) Treatment strategies for metastatic non- Lawrence TS, Chang EY, Hahn TM et al (1996) Radiosensi-
small-cell lung cancer. Clin Lung Cancer 1:34–41 tization of pancreatic cancer cells by 2’, 2’-difluoro-2’-
Johnson DH, Paul DM, Hande KR et al (1996) Paclitaxel plus deoxycytidine. Int J Radiat Oncol Biol Phys 34:867–872
carboplatin in advanced non-small-cell lung cancer: A Le Chevalier T, Brisgand D, Douillard JY et al (1994)
phase II trial. J Clin Oncol 14:2054–2060 Randomized study of vinorelbine and cisplatin versus
Kaplan B, Altýnbas M, Eroglu C, Karahacioglu E, Er O, Ozkan M, vindesine and cisplatin versusvinorelbine alone in advanced
Bilgin M, Canoz O, Gulmez I, Gulec M (2004) Preliminary non-small-cell lung cancer: results of a European multicen-
results of a phase II study of weekly paclitaxel (PTX) and ter trial including 612 patients. J Clin Oncol 12:360–367
carboplatin (CBDCA) administered concurrently with thoracic Lee JS, Scott C, Komaki R et al (1996) Concurrent chemora-
radiation therapy (TRT) followed by consolidation chemo- diation therapy with oral etoposide and cisplatin for locally
therapy with PTX/CBDCA for stage III unresectable non- advanced inoperable non-small-cell lung cancer: Radiation
small-cell lung cancer (NSCLC). Am J Clin Oncol 27(6): Therapy Oncology Group Protocol 91–06. J Clin Oncol
603–610 14:1055–1064
Kelly K, Chansky K, Gaspar LE, Albain KS, Jett J, Ung YC, Lee CB, Stinchcombe TE, Rosenman JG et al (2006) Thera-
Lau DH, Crowley JJ, Gandara DR (2008) Phase III trial of peutic advances in local-regional therapy for stage III non-
maintenance gefitinib or placebo after concurrent chemora- small-cell lung cancer: evolving role of dose-escalated
diotherapy and docetaxel consolidation in inoperable stage conformal (3-dimensional) radiation therapy. Clin Lung
III non-small-cell lung cancer: SWOG S0023. J Clin Oncol Cancer 8:195–202
26(15):2450–2456 Lee DH, Han JY, Cho KH, Pyo HR, Kim HY, Yoon SJ, Lee JS
Kim S, Kim M, Choi E et al (2007) Induction chemotherapy (2005) Phase II study of induction chemotherapy with
followed by concurrent chemoradiotherapy (CCRT) versus gemcitabine and vinorelbine followed by concurrent che-
CCRT alone for unresectable stage III non–small-cell lung moradiotherapy with oral etoposide and cisplatin in patients
cancer (NSCLC): Randomized phase III trial. J Clin Oncol with inoperable stage III non-small-cell lung cancer. Int J
25:391s (suppl; abstr 7528) Radiat Oncol Biol Phys 63(4):1037–1044
Klastersky J, Sculier JP, Lacroix H, Dabouis G, Bureau G, Leong SS, Fong KW, Lim WT, Toh CK, Yap SP, Hee SW,
Libert P, Richez M, Ravez P, Vandermoten G, Thiriaux J Tan EH (2010) A phase II trial of induction gemcitabine and
et al (1990) A randomized study comparing cisplatin or vinorelbine followed by concurrent vinorelbine and radio-
carboplatin with etoposide in patients with advanced non- therapy in locally advanced non-small cell lung cancer.
small-cell lung cancer: European Organization for Research Lung Cancer 67(3):325–329
and Treatment of Cancer Protocol 07861. J Clin Oncol Machtay M, Werner-Wasik M, DeNittis A et al. (2008) Pilot
8:1556–1562 study of carboplatin/radiotherapy plus ‘dose-dense’ pemetr-
Komaki R, Seiferheld W, Curran W et al (2003) Sequential exed for locally advanced non-small cell lung carcinoma.
versus concurrent chemotherapy, radiation therapy for J Clin Oncol. ASCO Annual Meeting Proceedings 26 7571
inoperable non-small cell lung cancer (NSCLC): analysis (abstract), 2008
of failures in a phase III study (RTOG 9410). Int J Radiat Manfredi JJ, Parness J, Horwitz SB (1982) Taxol binds to
Oncol Biol Phys 48:113 cellular microtubules. J Cell Biol 94:688–696
Kosmidis PA, Mylonakis N, Fountzilas G, Samantas E, Micetich KC, Barnes D, Ericson LC (1985) A comparative
Athanasiadis A, Andreopoulou E, Pavlidis N, Skarlos D study of the cytotoxicity of anticancer platinum drugs:
(1997) Paclitaxel and carboplatin in inoperable non-small- evidence that cis-diamminedichloroplatinum (II) and cis-
cell lung cancer: a phase II study. Ann Oncol 8:697–699 diammine-(l.I-cyclobutanedicarboxylato) platinum (II) dif-
Krzakowski M, Provencio M, Utracka-Hutka B, Villa E, fer only in the kinetics of their interaction with DNA.
Codes M, Kuten A, Henke M, Lopez M, Bell D, Biti G, Cancer Res 45:4043–4047
Merimsky O, Beorchia A, Riggi M, Caux NR, Pouget JC, Movsas B, Langer CJ, Ross HJ, Wang L, Jotte RM,
Dubray B, David P (2008) Oral vinorelbine and cisplatin Feigenberg S, Xu F, Huang CH, Monberg MJ, Obasaju CK
as induction chemotherapy and concomitant chemo-radio- (2010) Randomized phase II trial of cisplatin, etoposide, and
therapy in stage III non-small cell lung cancer: final radiation followed by gemcitabine alone or by combined
results of an international phase II trial. J Thorac Oncol gemcitabine and docetaxel in stage III A/B unresectable non-
3(9):994–1002 small cell lung cancer. J Thorac Oncol 5(5):673–679
Kudoh S, Takada M, Masuda N et al (1993) Enhanced Movsas B, Scott C, Sause W et al (1999) The benefit of
antitumor efficacy of a combination of CPT-11, a new treatment intensification is age and histology-dependent in
derivative of camptothecin, and cisplatin against human patients with locally advanced non-small cell lung cancer
lung tumor xenografts. Jpn J Cancer Res 84:203–207 (NSCLC): a quality-adjusted survival analysis of radiation
Langer CJ, Leighton JC, Comis RL et al (1995) Paclitaxel and therapy oncology group (RTOG) chemoradiation studies.
carboplatin in combination in the treatment of advanced Int J Radiat Oncol Biol Phys 45:1143–1149
non-small-cell lung cancer: A phase II toxicity, response, Movsas B et al (2003) Phase III study of amifostine in patients
and survival analysis. J Clin Oncol 13:1860–1870 with locally advanced non-small cell lung cancer (NSCLC)
Lau D, Leigh B, Gandara D (2001) Twice-weekly paclitaxel receiving chemotherapy & hyperfractionated radiation:
and weekly carboplatin with concurrent thoracic radiation Radiation Therapy Oncology Group (RTOG) 98–01. Pro
followed by carboplatin/paclitaxel consolidation for Stage Amer Soc Clin Oncol 22:636
III non-small-cell lung cancer: a california cancer con- Nakamura H, Yamaji Y, Fujita T et al (1995) Synergistic
sortium phase II trial. J Clin Oncol 19:442–447 growth inhibition of gemcitabine/cisplatin, gemcitabine/
310 R. Bland et al.

etoposide on a human lung cancer cell line, RERFLC-OK. Rich TA, Kirichenko AV (1998) Camptothecin radiation
Proc Am Assoc Cancer Res 36:A2425 sensitization: mechanisms, schedules, and timing. Oncology
Nitiss J, Wang JC (1988) DNA topoisomerase-targeting (Huntingt) 12(suppl 6):114–120
antitumor drugs can be studied in yeast. Proc Natl Acad Rosenman JG, Halle JS, Socinski MA et al (2002) High-dose
Sci U S A 85:7501–7505 conformal radiotherapy for treatment of stage IIIA/IIIB non-
Non-Small-Cell Lung Cancer Collaborative Group (1995) small-cell lung cancer: technical issues and results of a
Chemother- apy in non-small-cell lung cancer: a meta- phase I/II trial. Int J Radiat Oncol Biol Phys 54:348–356
analysis using updated data on individual patients from 52 Rowinsky EK, Donehower RC (1995) Paclitaxel (taxol).
randomized clinical trials. Br Med J 311:899–909 N Engl J Med 332:1004–1014
Omura M, Torigoe S, Kubota N (1997) SN-38, a metabolite of Rowinsky EK, Chaudhry V, Forastiere AA, Sartorius SE,
the camptothecin derivative CPT-11, potentiates the cyto- Ettinger DS, Grochow LB, Lubejko BG, Cornblath DR,
toxic effect of radiation in human colon adenocarcinoma Donehower RC (1993) Phase I and pharmacologic study of
cells grown as spheroids. Radiother Oncol 43:197–201 paclitaxel and cisplatin with granulocyte colony-stimulating
Parness J, Horwitz SB (1981) Taxol binds to polymerized factor: neuromuscular toxicity is dose-limiting. J Clin Oncol
tubulin in vitro. J Cell Biol 91:479–487 11:2010–2020
Patel SH, Ajlouni M, Chapman R, Lu M, Movsas B, Kim JH Rusthoven J, Eisenhauer E, Butts C et al (1999) Multitargeted
(2007) A prospective phase II study of induction carboplatin antifolate LY231514 as first-line chemotherapy for patients
and vinorelbine followed by concomitant topotecan and with advanced nonsmall cell lung cancer: a phase II study—
accelerated radiotherapy (ART) in locally advanced non-small National Cancer Institute of Canada Clinical Trials Group.
cell lung cancer (NSCLC). J Thorac Oncol 2(9):831–837 J Clin Oncol 17:1194–1199
Perez CA, Stanley K, Grundy G et al (1982) Impact of Rusu P, Ciuleanu TE, Cernea D, Pelau D, Gaal V, Cebotaru C,
irradiation technique and tumor extent in tumor control and Guttman T, Todor N, Ghilezan N (2007) Concurrent
survival of patients with unresectable non-oat cell carci- chemoradiotherapy with vinorelbine and a platinum com-
noma of the lung: report by The Radiation Therapy pound followed by consolidation chemotherapy for unre-
Oncology Group. Cancer 50:1091–1099 sectable stage III non-small cell lung cancer: preliminary
Perez CA, Pajak TF, Rubin P et al (1987) Long-term results of a phase II study. J BUON 12(1):33–39
observations of the patterns of failure in patients with Saka H, Shimokata K, Yoshida S et al (1997) Irinotecan (CPT-11)
unresectable non-oat cell carcinoma of the lung treated with and concurrent radiotherapy in locally advanced non-small
definitive radiotherapy: report by The Radiation Therapy cell lung cancer (NSCLC): a phase II study of Japan Clinical
Oncology Group. Cancer 59:1874–1881 Oncology Group (JCOG9504). Proc Am Soc Clin Oncol
Perol M, Guérin JC, Thomas P et al (1996) Multicenter 16:447a (Abstract 1607)
randomized trial comparing cisplatin-mitomycin-vinorel- Sakai H, Yoneda S, Kobayashi K, Komagata H, Kosaihira S,
bine versus cisplatin-mitomycinvindesine in advanced Kazumoto T, Saito Y (2004) Phase II study of bi-weekly
non-small cell lung cancer. Lung Cancer 14:119–134 docetaxel and carboplatin with concurrent thoracic radiation
Peterson P, Park K, Fossella F et al (2007) Is pemetrexed more therapy followed by consolidation chemotherapy with
effective in adenocarcinoma and large cell lung cancer than docetaxel plus carboplatin for stage III unresectable non-
in squamous cell carcinoma? A retrospective analysis of a small cell lung cancer. Lung Cancer 43(2):195–201
phase III trial of pemetrexed versus docetaxel in previously Sandler AB, Nemunaitis J, Denham C et al (2000) Phase III
treated patients with advanced non-small cell lung cancer trial of gemcitabine plus cisplatin versus cisplatin alone in
(NSCLC) 12th World Conference on Lung Cancer. J Thorac patients with locally advanced or metastatic non-small-cell
Oncol 2(suppl 4):S851 (Abstract P2-328) lung cancer. J Clin Oncol 18:122–130
Ratanatharathorn V, Lorvidhaya V, Maoleekoonpairoj S, Sause WT, Scott C, Taylor S et al (1995) Radiation Therapy
Phromratanapongse P, Sirilerttrakul S, Kraipiboon P, Oncology Group (RTOG) 88–08 and Eastern Cooperative
Cheirsilpa A, Tangkaratt S, Srimuninnimit V, Oncology Group (ECOG) 4588: preliminary results of a
Pattaranutaporn P (2001) Phase II trial of paclitaxel, carbo- phase III trial in regionally advanced unresectable non-
platin, and concurrent radiation therapy for locally advanced small-cell lung cancer. J Natl Cancer Inst 87:198–205
non-small-cell lung cancer. Lung Cancer 31:257–265 Scagliotti GV, Parikh P, von Pawel J et al (2008) Phase III study
Radiation Therapy Oncology Group. RTOG 0617/NCCTG comparing cisplatin plus gemcitabine with cisplatin plus
N0628/CALGB 30609 A randomzed phase III comparison pemetrexed in chemotherapy-naive patients with advanced-
of standard-dose (60 Gy) versus high dose (74 Gy) confor- stage non-small-cell lung cancer. J Clin Oncol 26:3543–3551
mal radiotherapy with concurrent and consolidation carbo- Schaake-Koning C, Van Den Bogaert W, Dalesio O et al (1992)
platin/paclitaxel in patients with stage IIIA/IIIB non-small Effects of concomitant cisplatin and radiotherapy on inoper-
cell lung cancer. http://www.rtog.org/members/protocols/ able non-small-cell lung cancer. N Engl J Med 326:521–530
0617/0617.pdf. Accessed 11 Mar 2011 Schiff PB, Fant J, Horwitz SB (1979) Promotion of microtubule
Reguart N, Viñolas N, Casas F, Gimferrer JM, Agustí C, assembly in vitro by Taxol. Nature 22:665–667
Molina R, Martin-Richard M, Sanchez-Reyes A, Gascón P Schild SE, McGinnis WL, Graham D et al (2006) Results of a
(2004) Integrating concurrent navelbine and cisplatin to phase I trial of concurrent chemotherapy and escalating
hyperfractionated radiotherapy in locally advanced non- doses of radiation for unrespectable non-small-cell lung
small cell lung cancer patients treated with induction and cancer. Int J Radiat Oncol Biol Phys 65:1106–1111
consolidation chemotherapy: feasibility and activity results. Seiwert T, Connell PP, Mauer AM et al (2007) A phase I study
Lung Cancer 45(1):67–75 of pemetrexed, carboplatin, and concurrent radiotherapy in
Radiotherapy and Third Generation Concurrent Chemotherapy Agents 311

patients with locally advanced or metastatic nonsmall cell Tishler RB, Geard CR, Hall EJ et al (1992a) Taxol sensitizes
lung cancer or esophageal cancer. Clin Cancer Res 13: human astrocytoma cells to radiation. Cancer Res
515–522 51:3459–3497
Segawa Y, Kiura K, Takigawa N, Kamei H, Harita S, Hiraki Tishler RB, Schiff PB, Geard CR et al (1992b) Taxol: a novel
S, Watanabe Y, Sugimoto K, Shibayama T, Yonei T, radiation sensitizer. Int J Radiat Oncol Biol Phys 22:
Ueoka H, Takemoto M, Kanazawa S, Takata I, Nogami 613–617
N, Hotta K, Hiraki A, Tabata M, Matsuo K, Tanimoto M Uejima H (2002) Dose-finding study of weekly irinotecan
(2010) Phase III trial comparing docetaxel and cisplatin (CPT-11) and carboplatin (CBDCA) with concurrent
combination chemotherapy with mitomycin, vindesine, thoracic radiotherapy (TRT) in locally advanced non-small
and cisplatin combination chemotherapy with concurrent cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 20:
thoracic radiotherapy in locally advanced non-small- A–A2699
cell lung cancer: OLCSG 0007. J Clin Oncol 28(20): Vafai D, Israel V, Zaretsky S et al (1995) Phase II trial of
3299–3306 combination carboplatin and taxol in non-small-cell lung
Senan S, Cardenal F, Vansteenkiste J, Stigt J, Akyol F, cancer (NSCLC). Proc Am Soc Clin Oncol 14:352
De Neve W, Bakker J, Dupont JM, Scagliotti GV, (abstr 1067)
Ricardi U, van Meerbeeck JP (2011) A randomized phase Vergnenègre A, Daniel C, Léna H, Fournel P, Kleisbauer JP,
II study comparing induction or consolidation chemother- Le Caer H, Letreut J, Paillotin D, Pérol M, Bouchaert E,
apy with cisplatin-docetaxel, plus radical concurrent che- Preux PM, Robinet G, Groupe Francais de Pneumo-
moradiotherapy with cisplatin-docetaxel, in patients with Cancerologie (2005) Docetaxel and concurrent radiotherapy
unresectable locally advanced non-small-cell lung cancer. after two cycles of induction chemotherapy with cisplatin
Ann Oncol 22(3):553–558 and vinorelbine in patients with locally advanced non-
Shewach DS, Hahn TM, Chang E et al (1994) Metabolism of small-cell lung cancer. A phase II trial conducted by the
2’, 2’-difluoro-2’-deoxycytidine and radiation sensitization Groupe Francais de Pneumo-Cancerologie (GFPC). Lung
of human colon carcinoma cells. Cancer Res 54:3218–3223 Cancer 47(3):395–404
Sinclair WK, Morton RA (1966) X-ray sensitivity during the Vokes EE, Herndon JE 2nd, Crawford J et al (2002) Random-
cell generation cycle of cultured Chinese hamster cells. ized phase II study of cisplatin with gemcitabine or
Radiat Res 29:450–474 paclitaxel or vinorelbine as induction chemotherapy fol-
Smit E, Mattson K, von Pawel J et al (2003) ALIMTA lowed by concomitant chemoradiotherapy for stage IIIB
(pemetrexed disodium) as second-line treatment of non- non-small-cell lung cancer: cancer and leukemia group B
small cell lung cancer: a phase II study. Ann Oncol 14: study 9431. J Clin Oncol 20:4191–4198
455–460 Vokes EE, Herndon JE II, Kelley MJ et al (2007) Induction
Socinski MA, Rosenman JG, Halle JS et al (2000) Induction chemotherapy followed by chemoradiotherapy compared
carboplatin/paclitaxel followed by concurrent carboplatin/ with chemoradiotherapy alone for regionally advanced
paclitaxel and dose-escalating conformal thoracic radiation unresectable stage III non–small-cell lung cancer: Cancer
therapy in unresectable stage IIIA/B non-small cell lung and leukemia group B. J Clin Oncol 25:1698–1704
cancer: a modified phase I trial. Cancer 89:534–542 Vokes EE, Senan S, Treat JA et al (2009) PROCLAIM: a
Socinski MA, Blackstock AW, Bogart JA et al (2008) phase III study of pemetrexed, cisplatin, and radiation
Randomized phase II trial of induction chemotherapy therapy followed by consolidation pemetrexed versus
followed by concurrent chemotherapy and dose-escalated etoposide, cisplatin, and radiation therapy followed by
thoracic conformal radiotherapy (74 Gy) in stage III non- consolidation cytotoxic chemotherapy of choice in locally
small-cell lung cancer: CALGB 30105. J Clin Oncol advanced stage III non small-cell lung cancer of other than
26:2457–2463 predominantly squamous cell histology. Clin Lung Cancer
Solomon B, Bunn PA Jr (2005) Clinical activity of pemetrexed: 10:193–198
amultitargeted antifo- late anticancer agent. Future Oncol Wasserman T, Chapman JD (2004) Radiation response
1:733–746 modulation. Part A: chemical sensitizers and protectors.
Solomon B, Ball DL, Richardson G, Smith JG, Millward M, In: Perez C, Brady L, Halperin E et al (eds) Principles,
MacManus M, Michael M, Wirth A, O’Kane C, practice of radiation oncology, 4th edn. JB Lippincott,
Muceniekas L, Ryan G, Rischin D (2003) Phase I/II study Philadelphia, PA
of concurrent twice-weekly paclitaxel and weekly cisplatin Wozniak AJ, Crowley JJ, Balcerzak SP et al (1998) Random-
with radiation therapy for stage III non-small cell lung ized trial comparing cisplatin with cisplatin plus vinorelbine
cancer. Lung Cancer 41(3):353–361 in the treatment of advanced non-small-cell lung cancer:
Stern A, Sevin BU, Perras J et al (1993) Taxol sensitizes human a Southwest Oncology Group study. J Clin Oncol 16:
ovarian cancer cells to radiation. Cancer Res 48:252–258 2459–2465
Takeda K, Negoro S, Kudoh S et al (1999) Phase I/II study of Xu Y, Ma S, Ji Y et al (2010) Concomitant chemoradiotherapy
weekly irinotecan and concurrent radiation therapy for using pemetrexed and carboplatin for unresectable stage III
locally advanced non-small cell lung cancer. Br J Cancer non-small cell lung cancer (NSCLC): Preliminary results of
79:1462–1467 a phase II study. Lung Cancer. (Epub ahead of print)
Tanzer LR, Rutherford PG, Self TD et al (1995) Antitumor Yamada M, Kudoh S, Fukuda H et al (2002) Dose-escalation
activity of gemcitabine in combination with cisplatin study of weekly irinotecan and daily carboplatin with
against the human NSCLC xenograft Calu-6. Proc Am concurrent thoracic radiotherapy for unrespectable stage III
Assoc Cancer Res 36:A1761 non-small cell lung cancer. Br J Cancer 87:258–263
312 R. Bland et al.

Yamamoto N, Nakagawa K, Nishimura Y et al (2010) Phase III Zatloukal P et al (2004) Concurrent versus sequential chemo-
study comparing second- and third-generation regimens radiotherapy with cisplatin and vinorelbine in locally
with concurrent thoracic radiotherapy in patients with advanced non-small cell lung cancer: a randomized study.
unresectable stage III non-small-cell lung cancer: West Lung Cancer 46(1):87–98
Japan Thoracic Oncology Group WJTOG0105. J Clin Oncol
28:3739–3745
Conventional Radiation Therapy in Early
Stage Non-small-cell Lung Cancer
Branislav Jeremić, Sinisa Stanic, and Slobodan Milisavljevic

Contents Abstract
Surgery is standard treatment approach in patients
1 Introduction.............................................................. 315 with early stage (I-II) nonsmall cell lung cancer.
2 Overall Results of Radiation Therapy .................. 316 However, there are patients who do not undergo
surgery due to existing comorbidities, advanced
3 Tumor Dose .............................................................. 319
age or refusal. They have traditionally been treated
4 Tumor Stage and Size............................................. 322 with radiation therapy which provided median
5 Treatment Volume................................................... 323 survival times of [ 30 months and 5-year survival
6 Prognostic Factors ................................................... 327
rates of [ 30% in stage I disease.

7 Toxicity...................................................................... 328
8 Quality of Life.......................................................... 331
1 Introduction
9 Novel Approaches .................................................... 332
9.1 Stereotactic Radiation Therapy ................................. 332
9.2 Accelerated Hypofractionated Radiation
Surgery is considered a worldwide standard treatment
Therapy ...................................................................... 333 approach in patients with early stage (I/II) non-small-
9.3 Accelerated Hyperfractionated Radiation Therapy cell lung cancer. It offers the best chance for cure and
and Concurrent Radiation Therapy results did not change substantially in the past three
with Chemotherapy ................................................... 334
decades. In the International staging system classifi-
10 Conclusions ............................................................... 334 cation (Mountain 1986), the 5-year survival rates for
References.......................................................................... 335 pathological stage I/II were 68.5% for T1N0, 59% for
T2N0, 54.1% for T1N1 and 40.0% for T2N1. When
clinical staging is used, however, these results became
inferior: 61.9% for T1N0, 35.8% for T2N0, 33.6% for
T1N1, and 22.7% for T2N1 tumors. A similar analysis
was done for the purpose of the second and then third
staging classification (Mountain 1997; Goldstraw et al.
B. Jeremić (&) 2007). According to the last staging classification
Institute of Lung Disease, Sremska Kamenica, Serbia (Goldstraw et al. 2007). Stage I now has subdivision to
e-mail: nebareje@gmail.com IA (T1a, b N0) and IB (T2a N0), while the stage II also
S. Stanic, has two subdivisions, to IIA (T1a, b, T2a N1, T2b N0)
Department of Radiation Oncology, and IIB (T2b N1 and T3 N0).
University of California Davis, Sacramento, USA
The surgical/pathological staging is considered an
S. Milisavljevic ultimate one, giving the best correlation with the out-
Department of Thoracic Surgery, University Hospital,
come. There are, however, also data from surgical series
Kragujevac, Serbia

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 315
DOI: 10.1007/174_2011_207, Ó Springer-Verlag Berlin Heidelberg 2011
316 B. Jeremić et al.

on patients with early stage nonsmall cell lung cancer only, serious flaws of that report, both methodological
when clinical staging is used (Mountain 1986; Naruke and statistical, led to the raise of the concern (Jeremic
et al. 1988). Although comparison of treatment outcome et al. 2003b) that observation-only should not be prac-
based on pathological and clinical staging is not so fre- ticed in any case with early stage non-small-cell lung
quently observed in recent years, nevertheless, there is cancer today. More recently, (Raz et al. 2007a, 2007b)
still a subset of patients in whom clinical stage is used, reviewed the California Cancer Center registry and
irrespective of treatment intention. Data from the sur- reported on a median survival of only 9 months in 1,432
veillance end results suggest that as many as 31% of all patients with stage I non-small-cell lung cancer that
patients with localized non-small-cell lung cancer do not received no initial treatment. Indirect evidence support-
undergo lobectomy (Potosky et al. 2004). Majority of ing active treatment also came from the recent study
these patients, although technically resectable, are not (Henschke et al. 2003) which showed that even smallest
undergoing surgery due to various reasons. The vast tumors (i.e., stage I) when untreated had 8-year fatality
majority of these patients are medically inoperable due rate of 87, 94, and 88%, for tumors having sizes of 6–15,
to pre-existing comorbidity, mostly cardiopulmonary, 16–25 and 26–30 mm, respectively. Although that study
presumably carrying high peri- and post-operative risk. focused on the role of surgery versus observation, it
Another group of patients not undergoing surgery are would not be unreasonable to expect the same or similar
elderly, owing to presumed restricted cardiopulmonary if the radiation therapy alone had been active treatment in
reserve, expected to occur even without overt cardio- a study similar to this. This is especially so when one
pulmonary disease. Finally, the smallest group of considers excellent results obtained in the last decade
patients not undergoing surgery and possibly the group with the use of stereotactic radiation therapy in early
of patients having the major importance for radiation stage non-small-cell lung cancer. Although stereotactic
oncologist are the patients who refuse surgery. This may body radiation therapy is being increasingly used for
nowadays range from as low as nil to as high as[20% in early stage medically inoperable non-small-cell lung
elderly treated with pneumonectomy (Whittle et al. cancer, conventionally fractionated, three-dimensional
1991; Au et al. 1994; Mizushima et al. 1997). These conformal radiation therapy is still used in radiation
three groups of patients are those mostly offered radia- oncology clinics since it does not require strong physics
tion therapy alone, being frequently considered ‘‘stan- support for a very rigorous quality assurance and pre-
dard’’ treatment approach in this setting. Unfortunately, cise immobilization, which cannot be tolerated by all
the patients who undergo radiation therapy alone for patients.
early stage nonsmall cell lung cancer mostly constitute This chapter, therefore, summarizes achievements
negative selection, materialized in their serious con- of conventional radiation therapy in early stage non-
comitant diseases, the use of clinical staging, and small-cell lung cancer. It aims to highlight its
insufficient staging as well. It is, therefore, quite clear advantages and underline its disadvantages, and also
that the results of radiation therapy in this population enable better insight in a number of pre-treatment and
cannot be meaningfully compared to those of surgery, treatment characteristics in this setting, focusing on
even when one use the results from the surgical series curative radiation therapy.
using clinical staging. Additionally, there is always a
bias in reporting radiation therapy versus surgical series
as well as institutional/investigator bias. Finally, differ- 2 Overall Results of Radiation
ent process of decision-making (patients vs. physicians) Therapy
has to be accounted for, due to great variance observed
across the studies with the respect to the proportion of Numerous studies unequivocally documented the out-
patients refusing surgery. come of patients with operable non-small-cell lung
There are no prospective randomized studies com- cancer in the last four decades, including mostly
paring radiation therapy alone with other treatment patients with early (I/II) stage disease. The history of
modalities in patients with early stage non-small-cell radiation therapy in early stage non-small-cell lung
lung caner, including observation (no treatment). In spite cancer started with the report of Morrison et al. (1963)
of the fact that one report (McGarry et al. 2002) showed on 28 patients with ‘‘operable’’ lung cancer treated with
no advantage for radiation therapy over observation- 45 Gy who achieved the overall survival of 7% at
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 317

5 years. While one may express disappointment with study of Fang et al. (2006) who evaluated the outcome of
these early and clearly inferior results (comparing to 200 patients with medically inoperable stage I non-
nowadays standards and achievements), this should small-cell lung cancer and compared three-dimensional
come as no surprise due to the lack of modern diag- with two-dimensional radiation therapy planning and
nostic and planning tools (computerised tomography). execution. The overall survival, disease-specific sur-
Similarly Coy and Kennelly (1980) provided results not vival, and locoregional control rates were significantly
very different from those of Morrison et al. (1963). higher in patients treated with three-dimensional radia-
They have achieved 5-year survival of 10% in 141 tion therapy. Two- and 5-year overall survival rates were
patients with T1-3 NX tumors treated with radiation 68, 36, 47 and 10%, respectively (p = 0.001). This was
therapy doses of 50–57.5 Gy. It was Smart (1966) who confirmed by a multivariate analysis using overall sur-
was the very first to indicate greater potential for radi- vival and disease-specific survival. However, not all
ation therapy alone in this disease. Using total radiation investigators reported on the similar experience. In the
therapy doses of 40–55 Gy in 40 patients he obtained a study of Lagerwaard et al. (2002), the 5-year overall
5-year survival of 22% and the median survival time of survival was only 12% which led authors to conclude
approximately 24 months. Of the studies that followed that three-dimensional radiation therapy did not improve
later on (Cooper et al. 1985; Haffty et al. 1988; Noordijk survival. Possible explanation for inferior results of
et al. 1988; Zhang et al. 1989; Talton et al. 1990; Lagerwaard et al. (2002) could include as much as one-
Sandler et al. 1990; Ono et al. 1991; Dosoretz et al. third of patients having a history of a previous malig-
1992; Kupelian et al. 1996; Cheung et al. 2000) some nancy, smaller percentage of those refusing surgery,
enrolled patients without specifying results according larger proportion of those having T2 tumors as well as
to the tumor stage, while others also enrolled a pro- the lack of confirmatory pathology in as much as 40% of
portion of patients in stage III NSCLC (Cooper et al. patients. One additional study (Bradley et al. 2003)
1985; Zhang et al. 1989; Talton et al. 1990; Dosoretz failed to show expected advantage of three-dimensional
et al. 1992; Kupelian et al. 1996; Cheung et al. 2000). conformal radiation therapy, likely as a consequence of
As expected, there was a substantial variation in the unfavorable patient characteristics. As much as 60% of
diagnostic tools used over the time and in the first their patients had greater than 5% weight loss and almost
reports computerised tomography scanning for diag- 40% of patients received elective nodal irradiation that
nostic and therapeutic (planning) purposes was not may have caused toxicities that led to poor outcome.
used. Therefore, studies covering longer periods of Radiation therapy characteristics greatly varied
time were more likely to include a number of patients with the time, too. Doses as low as 18 Gy were
with more (locoregionally) advanced disease sometimes given, but went up to 80 Gy, while virtu-
(Morrison et al. 1963; Smart 1966; Coy and Kennelly ally all fractionation regimens were used: standard
1980; Cooper et al. 1985; Haffty et al. 1988; Noordijk (1.8–2.0 Gy per fraction), hypofractionated (up to
et al. 1988; Zhang et al. 1989; Talton et al. 1990; 4 Gy per fraction), split-course (one or two weeks
Sandler et al. 1990; Ono et al. 1991; Dosoretz et al. split), or hyperfractionated (1.2 Gy b.i.d. fraction-
1992; Kupelian et al. 1996; Cheung et al. 2000). To ation). Treatment machines used to deliver irradiation
further extend this, even some of the reports published in also ranged from orthovoltage X-rays through cobalt-
the eighties and the nineties of the last century suffered 60 to either low- or high-megavoltage X-rays of linear
from the very same matter (Cooper et al. 1985; Zhang accelerators, as well as did treatment prescription/
et al. 1989; Talton et al. 1990; Haffty et al. 1988; dose specification, patients positioning, number of
Noordijk et al. 1988; Sandler et al. 1990; Ono et al. irradiated treatment fields per day, etc.
1991). This may be one of the crucial issues in inter- Whatever the differences and variances in the
pretation of the overall results, since Sandler et al. (1990) aforementioned studies and the interpretation of their
documented an improvement in survival of patients with respective results may have led to, when the data from
‘‘excellent’’ staging (chest CT scan, including the liver, the literature were summarized (Jeremic et al. 2002)
and bone scan) when compared to those having ‘‘good’’ they showed that radiation therapy alone was capable
staging (conventional tomography, liver–spleen scan, of producing a median survival time of [30 months
and a bone scan), and particularly to those being staged ([40 months in T1N0) since the mid-eighties of the
less vigorously. Confirmation for this came with recent last century, with 5-year survival rates of [30% in
318 B. Jeremić et al.

Fig. 1 CT of the chest in a 67-year old female with medically following completion of radiation therapy. Note a complete
inoperable, T1bN0M0, peripherally located, non-small-cell response to conventionally fractionated radiotherapy. Three
lung cancer of the left lower lobe, status post 3D conformal years after radiation therapy, the patient is alive and without
radiotherapy to a total dose of 64 Gy in 2 Gy fractions. a CT evidence of locoregional disease recurrence or distant
prior to radiation therapy; b CT of the same patient nine months metastases

stage I non-small-cell lung cancer (40% in T1N0) and (Jeremic et al. 2005). Interestingly, the highest med-
up to 25% in stage II non-small-cell lung cancer ian survival times (up to 36 months) and the highest
(Fig. 1a, b). The evidence that came from the avail- 5-year survival rates (up to 35%) were observed in
able studies published in the last decade reconfirms these particular studies. It became widely accepted
these statements (Fang et al. 2006; Zhao et al. 2007; opinion that these patients represent the population
Low et al. 2007; Hsie et al. 2007; Pemberton et al. which seems to be the one most likely to give true
2009; Sandhu et al. 2009; Campeau et al. 2009; insight in the effectiveness of radiation therapy in this
Newlin et al. 2009; Watkins et al. 2010), while pre- disease, simply because they are those resembling
treatment and treatment characteristics (available surgical candidates at most. This is also observed in
histology, staging, imaging, three-dimensional plan- studies using stereotactic radiation therapy in early
ning based on CT scanning) became always available stage non-small-cell lung cancer. Reporting on overall
and, except time-dose-fractionation and the dose survival as an endpoint in this patient population
prescription) almost uniform. becomes more scientifically meaningful, due to less
Not only the differences in radiation therapy cancer-unrelated events occurring. In other patient
characteristics in the aforementioned studies exist, but populations (medically inoperable, elderly), the use of
these results were also achieved in a cohort of sub- cancer-specific survival or disease-specific survivals
stantially differing patient populations. An important must be mandatory to correct for events other than
underlying issue, namely, the reason for not under- cancer-related. Indeed, when 5-year cancer-specific or
going surgery, was recently recognized as one of the disease-free survival rates were reported (Sandler
major contributors to existing differences between et al. 1990; Kaskowitz et al. 1993; Slotman and Karim
studies, including their outcomes. It was considerably 1994; Krol et al. 1996; Sibley et al. 1998; Cheung
different across the studies, particularly when one et al. 2000; Zhao et al. 2007), they were usually twice
consider patient refusal which only recently started to as high as those of overall survival, as presented in the
gain more attention. While the percentage of such same studies, the difference being approximately
patients was usually around 10%, there are also sev- 10–20% in favor of the former. Additionally, it is well
eral studies in which it was[20% (Zhang et al. 1989; recognized fact that patients’ refusal inversely corre-
Morita et al. 1997; Jeremic et al. 1997, 1999; Fang lates with the incidence of intercurrent deaths
et al. 2006), and in one study it was even 70% (6–16%) (Zhang et al. 1989; Sandler et al. 1990;
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 319

Jeremic et al. 1997, 1999). The incidence of the Dose–response relationship was evaluated by
intercurrent deaths, on the other side, directly many investigators. It has usually been observed that
dependent on the increasing age and pre-existing higher doses carry favorable outcome. Some studies
comorbidity (21–43%) (Noordijk et al. 1988; used somewhat lower cut-off values (e.g., 40 or
Kaskowitz et al. 1993; Slotman and Karim 1994; 50 Gy) which enabled comparison of palliative versus
Morita et al. 1997; Sibley et al. 1998; Zhao et al. curative treatments (Cooper et al. 1985; Sandler et al.
2007). These important facts form a complicated 1990; Kupelian et al. 1996). It was Cooper et al.
framework which has largely been underestimated in (1985) who first noted improved survival with the
the past. Contemporary studies must take these facts higher dose ([40 Gy compared to \40 Gy). Haffty
into account and adapt the study designs and data et al. (1988) noted an advantage of continuous course
presentation so as to enable better insight into the (59 Gy) over split-course (54 Gy) regimen, not only
effectiveness of radiation therapy in this disease and regarding overall survival, but local control as well.
to enable easier comparison between the studies. The dose–effect upon survival was also evaluated in
Irrespective of these shortcomings, results of radi- the study of Zhang et al. (1989) who found that higher
ation therapy in recent years were occasionally com- doses (69–70 Gy) were more efficient than the lower
pared to those of surgery. While this matter will be a ones (55–61 Gy). However, Sandler et al. (1990)
subject of discussion in other chapters, here only a brief could not confirm this, presumably due to a somewhat
emphasis will be made. In such one study, Hsie et al. narrow dose range in their study Hayakawa et al.
(2007) provided a single-institutional comparison of (1992) and Dosoretz et al. (1992) also confirmed
outcomes of limited resection and radical radiation importance of higher doses on overall survival and
therapy. Radiation therapy characteristics included a disease-specific survival, but warned on the use of
CT-planned and delivered median radiation therapy very high doses ([80 Gy) when conventional tools
doses of 70 Gy (range, 60–75 Gy) and the median daily were used for treatment planning/delivery (Hayakawa
fraction size of 2.5 Gy (range, 2.0–4.11 Gy). The et al. 1992) due to increased risk of treatment-related
3-year actuarial survival for patients treated with lim- toxicity and mortality Slotman and Karim (1994) did
ited resection was 62.7% and median survival was not not find an impact of the higher (48–56 Gy) versus
reached. The actuarial 3-year survival for patients lower (32–40 Gy) doses of radiation therapy on either
treated with radiation therapy was 55% and median overall survival or disease-specific survival in stage I
survival was 38 months (n.s.). non-small-cell lung cancer. Kaskowitz et al. (1993)
and Sibley et al. (1998) both observed better overall
survival, though not statistically significant, for higher
doses ([65 and [64 Gy, respectively). Graham et al.
3 Tumor Dose (1995) used tumor/dose/fractionation calculations as a
measure of the effectiveness of radiation therapy to
To start properly addressing the question of the document better outcome with increasing tumor/dose/
effectiveness of radiation therapy in early stage fractionation values in multivariate analysis. Also,
non-small-cell lung cancer, one must start with the Kupelian et al. (1996) and Morita et al. (1997)
radiation therapy dose itself. Data from the literature showed impact of the dose on local response/control,
identifies tumor doses used during the radiation which was not always translated into a better survival
therapy course ranging from as low as 18 Gy to as (Morita et al. 1997). Stage I/II non-small-cell lung
high as 90 Gy or as high as biologically equivalent cancer should represent tumors with the smallest
dose of 124.5 Gy. While this range of tumor doses burden of tumor cells, although imprecise staging
used should somehow enable getting information currently used (still based on tumor size rather than on
regarding the anticipated dose–response (effect) issue, the volume) allows that even small-volume tumors
it cannot artificially be detached from the issue of are placed in the higher staging category (e.g., stage
tumor stage/size, since one of the long-lasting bio- III) when invading certain intrathoracic and/or
logical premises in radiation oncology is that larger mediastinal structures. While some of the existing
tumor sizes (presumably higher stage) require higher analyses favored even lower doses of radiation
tumor doses. therapy, it still remains preferable to use the doses
320 B. Jeremić et al.

traditionally considered as ‘‘curative’’, being in the One should also take into consideration altered
order of [65–70 Gy with standard fractionation or its fractionated regimens. Beside studies Jeremic et al.
equivalent when altered fractionation is used. This (1997, 1999) who successfully used hyperfractionated
suggestion should be even more valid nowadays with radiation therapy (69.6 Gy using 1.2 Gy b.i.d.) in
widespread of three-dimensional treatment planning both stage I and II non-small-cell lung cancer,
and delivery, because it should enable better thera- accelerated regimens were also used, such as contin-
peutic benefit when compared to two-dimensionally uous hyperfractionated accelerated radiation therapy
planned and executed radiation therapy used in the (Saunders et al. 1999a). Recent report of Pemberton
past. Indeed, the data on the use of high-dose radia- et al. (2009) not only reconfirmed its effectiveness,
tion therapy (e.g., 80 Gy) show that it may indeed be but also brought to the light effectiveness of another
very beneficial. In the study of Watkins et al. (2010) hypofractionated regimen, the one using 55 Gy in 20
100 primary lung tumors in 98 patients of a clinical daily fractions. Both regimens are almost exclusively
stage I and II were treated with C70 Gy at 1.8– practiced in the UK. In that study, 137 patients treated
2.5 Gy. The median prescribed dose was 80 Gy with CHART and 140 patients treated with 55 Gy
(range, 70–90 Gy) with 84% of patients receiving in 20 daily fractions were compared. The median
[79 Gy. The calculated biologically equivalent dose survivals were 16.6 and 21.4 months, while 2-year
(at 2 Gy per fraction with alpha/beta estimated at survivals were 34 and 45% for the two regimens,
10 Gy) was [95 Gy for 80 patients, and [99 Gy respectively. No formal comparison was provided in
for 50 patients. The estimated 2- and 3-year local this study, although provided statistical analysis
(in-field) controls were 53 and 50%, respectively, clearly indicated superiority of 55 in 20 daily frac-
while estimated 2- and 3-year overall survivals were tions over CHART.
47 and 24%, respectively. Bradley et al. (2005) performed a phase I–II dose-
Also, Zhao et al. (2007) reported on the study escalation study using three-dimensional conformal
showing that high radiation dose may reduce the radiation therapy in 177 patients with medically
negative effect of large gross tumor volume in inoperable stage I–III non-small-cell lung cancer
patients with medically inoperable stage I–II non- (RTOG 9311). Concurrent chemotherapy with radia-
small-cell lung cancer, In that study, multivariate tion was not allowed. The radiation dose was safely
analysis showed that there was a significant interac- escalated to 83.8 Gy for patients with lung V20\25%
tion between radiation dose and gross tumor volume and 77.4 Gy for patients with lung V20 between 25
(p \ 0.001). In patients with biologically equivalent and 36% utilizing fraction sizes of 2.15 Gy. The
dose of B79.2 Gy, the overall survival medians for highest dose level, 90.3 Gy was too toxic, resulting in
patients with gross tumor volume of [51.8 cm3 and dose-related deaths in 2 patients. Less than 10% of
B51.8 cm3 were 18.2 and 23.9 months, respectively patients received elective nodal irradiation. Locore-
(p = 0.015). If biologically equivalent dose was gional control, which was a secondary objective of
[79.2 Gy, no significant difference was found this dose escalation study, was achieved in 50–78% of
between two gross tumor volume groups. For patients.
patients with gross tumor volume [51.8 cm3, the At the present time, different dose fractionation
overall survival; medians in those with biologically schedules have been used in radiation oncology.
equivalent dose of [79.2 Gy and \79.2 Gy were Doses have ranged between 60 and 70 Gy (Fig. 2). If
30.4 and 18.2 months, respectively (p \ 0.001). If radiation therapy is delivered in once daily fractions,
gross tumor volume was B51.8 cm3, the difference five days per week, 1.8–2.0 Gy daily fractions are a
was no longer significant. Similarly, in the study of reasonable approach. In the United States, if stereo-
Low et al. (2007) in 23 patients with stage I non- tactic body radiation therapy is not available in a
small-cell lung cancer, the median survival for radiation oncology clinic, or a patient is unable to
patients who received a biologically equivalent dose tolerate stereotactic immobilization, conventionally
of C63.9 Gy was not reached as compared to fractionated radiation therapy utilizing three-dimen-
20 months in patients with biologically equivalent sional conformal planning is considered, if possible to
dose of \63.9 Gy (p = 0.03). a total dose of 70 Gy with 2 Gy per fraction and the
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 321

Fig. 2 3D conformal radiotherapy treatment plan for a 69-year Heterogeneity correction was applied to take into account the
old male with medically inoperable, T1bN0M0, peripherally density differences. Note the dose distribution and volumes:
located, non-small-cell lung cancer of the right upper lobe, GTV = gross tumor volume, CTV = clinical target volume and
status post CT guided fine needle aspiration. A dose of 70 Gy PTV = planning target volume
was delivered in 2 Gy fractions with five conformal fields.

goal to keep total lung volume receiving 20 Gy (V20) 3.5–4.0 Gy per fraction, to a total dose of 48–60 Gy
\35%. One of the disadvantages of conventional (Soliman et al. 2011). This is not a type of stereotactic
fractionation is a long treatment course, 6–7 weeks. body radiation therapy, but rather an alternative to
In order to reduce the number of treatments required extremely hypofractionated stereotactic radiation.
to complete a definitive course of radiation therapy, a This approach will be presented later in this chapter
new approach has emerged—accelerated hypofrac- when we discuss novel approaches in radiation ther-
tionated radiation therapy, which typically employs apy for early stage non-small-cell lung cancer.
322 B. Jeremić et al.

providing causal relationship between local control


4 Tumor Stage and Size and overall survival. Therefore, local/regional-recur-
rence free survival or disease-specific survival must
When influence of tumor stage and/or size on treat- be included in the analysis as important initial end-
ment outcome was evaluated, it was mostly observed points. The distant metastasis-free survival must be
that smaller tumors and/or lower stage of the disease used, too, to provide better insight into the events
carry an improvement in survival. Different cut-off other than those occurring locoregionally. The pat-
sizes were used in various studies, but sizes most terns of failure (detailed later in the chapter) were
frequently used were \3 cm or \4 cm and these shown to heavily depend on local/regional tumor
tumors were frequently compared to larger ones. control in this disease. In one such analysis, Watkins
Also, T1 stage was frequently compared to T2 stage et al. (2010) showed that results were significantly
with regard to overall survival, disease-specific sur- better for cT1N0 over cT2N0 tumors using estimated
vival and local control. Multivariate analyses were 2- and 3-year local control (66 vs. 42%), and disease-
also used to in an effort to confirm independent free survivals (48 vs. 26%).
influence of T stage, frequently documenting that Important obstacles for clear and precise definition
T stage was the only prognosticator of the treatment of the role of tumor stage/size are staging systems
outcome (Kaskowitz et al. 1993; Graham et al. 1995; widely used in the last twenty years (Mountain 1986,
Gauden et al. 1995; Jeremic et al. 1997; Fang et al. 1997; Goldstraw et al. 2007). As already mentioned,
2006). Additionally, better outcome for T1 versus T2 these surgical systems do not relate only to a tumor
or for tumor size B3 cm versus [3 cm (Morita et al. size, but also to a particular tumor location, leading to
1997; Slotman et al. 1996; Sibley et al. 1998; Yu et al. confusion when this (surgical) staging system is
2008) was observed, although without statistical sig- applied to non-surgical setting, having different bio-
nificance. Contrary to these, there were also studies logical premises. Practical example of such a problem
that evaluated both T stage and particular tumor size, is as follows: tumor of 1 cm would be, by virtue of its
with conflicting results. In the study of Kupelian et al. size placed into T1 category, but if it involves main
(1996) T stage did not influence either overall sur- bronchus at C2 cm distal to the carina, it would be
vival or disease-specific survival or local control. designated as T2. This may be even more so in the
Interestingly, however, when tumor size was used as a case of a tumor of the same size invading chest wall,
variable, it was found that tumors \5 cm had better being automatically placed into the T3 category. This
disease-specific survival and those \4 cm had better issue may be an important one owing to the log-cell
local control, confirmed in both cases using multi- kill nature of anticancer action of radiation therapy.
variate analysis. Zhao et al. (2007) also showed There are requests for continuous revision of current
superior outcome for patients with stage I versus international staging system, which should make both
those with stage II in multivariate analysis in that T and N staging more specific/detailed, computer-
study, as well as did Pemberton et al. (2009). In that enabled measurements of T and N volumes enabled
study, gross tumor volume was used as variable. and, therefore, efforts/results are easier to interpret/
Several studies also demonstrated negative effect of compare. Indeed, several aforementioned studies have
increasing gross tumor volumes on treatment out- successfully shown that gross tumor volume can be
comes for radiation therapy in non-small-cell lung used as important predictor of treatment outcome.
cancer (Basaki et al. 2006; Bradley et al. 2002; Positron emission tomography (PET) has become a
De Petris et al. 2005; Etiz et al. 2002 Yu et al. 2008). standard modality for staging non-small-cell lung
Contrary to these, Willner et al. (2002) reported that cancer (Lin and Alavi 2009). With regard to intra-
for tumors larger than 100 cm3, no dose effect was thoracic nodal staging, PET has a definite role in
seen. In their study, only two out of 45 tumors [100 communities in which mediastinoscopy is not avail-
cm3 were controlled for [2 years in patients with able, whereas the impact can be quite limited in
various stages of the disease. While one may expect institutions in which invasive mediastinal staging is
impact of tumor stage/size on the outcome, it is rea- available. Although the role of PET scanning in
sonable to expect that this would happen first at local/ staging will be discussed in a separate chapter, two
regional level, and then on the overall survival, questions seem as important ones. The first question is
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 323

whether positive PET scans in the mediastinum To better address this issue, one must consider it
require mediastinoscopy. Since the false-positive rate together with the irradiation dose used for elective
of PET in the mediastinum is 15–20% (Detterbeck treatment. Some studies used doses of 40 Gy in 20
et al. 2004), most physicians agree that positive daily fractions (Zhang et al. 1989; Talton et al. 1990;
results on PET scan should be confirmed by a biopsy Hayakawa et al. 1992; Slotman et al. 1996; Morita
and mediastinoscopy should be recommended. The et al. 1997; Hayakawa et al. 1999) which can not be
second question is whether negative PET scans in the considered as adequate to treat microscopic disease.
mediastinum obviate the need for mediastinoscopy. While some were using 45 Gy in 20–22 fractions
It is still controversial whether or not to confirm a (Morrison et al. 1963; Haffty et al. 1988; Kupelian
negative PET scan result in the mediastinum by et al. 1996) which can be considered as standard of
mediastinoscopy. If false-negative rate of PET in the practice, it is of unproven efficacy in lung cancer.
mediastinum is only 5–8% (Detterbeck et al. 2004), Additionally, if we extrapolate the data from squa-
mediastinoscopy can potentially be avoided. Some mous-cell carcinoma of the head and neck, than one
physicians would argue that this false-negative rate is would need 50 Gy given with 2.0 Gy standard frac-
low enough to be acceptable while others would tionation to treat microscopic disease successfully.
debate that one should not rely on a negative PET Close to this level were doses used by Kaskowitz
study, particularly in patients with central tumors, et al. (1993) by Morita et al. (1997) in part of their
adenocarcinoma histology, and N1 nodal involvement patients, and by Jeremic et al. (1997, 1999), with
by CT imaging. Cerfolio et al. (2006) recommended hyperfractionated radiation therapy dose of 50.40 Gy
consideration of mediastinoscopy in PET negative using 1.2 Gy b.i.d. fractionation applied by the latter.
patients with adenocarcinoma, upper lobe tumors, or Finally, because of the fear that there may be an
tumors with maximum SUV C10. Presently, in the increased risk of subclinical nodal spread in some
United States, many believe that negative PET scan in lymph node regions, others have also adapted other-
the mediastinum obviate the need for mediastinos- wise strict institutional policy and included some
copy only in patients with T1 peripheral tumors, nodes at risk into the limited field radiation therapy,
while for all other cases mediastinoscopy should be giving it, therefore, a form of ‘‘electively-limited’’
strongly considered. Radiation oncologists, however, radiation therapy, usually based on primary tumor
often face another challenge: the majority of medi- location (central tumor location or tumor adjacent to
cally inoperable patients with early stage non-small- the mediastinum (Senan et al. 2002; Lagerwaard et al.
cell lung cancer decline mediastinoscopy, as an 2002).
invasive diagnostic procedure. In this situation, radi- However, the choice and the number of the treat-
ation oncologists have no other choice than to use ment fields and the dose prescription was not always
clinical staging by PET/CT for treatment volume clearly specified, causing somewhat less precise
definition. interpretation of the data. This is so particularly
regarding the second part of the radiation therapy
course, which, by using various combinations of
5 Treatment Volume radiation therapy fields to treat visible tumor only
(mostly obliques and/or laterals), provides an unin-
Another issue of particular importance in the field of tentional treatment contribution to the nodal areas at
radiation therapy of lung cancer is the ‘‘optimal’’ risk. This contribution for a particular radiation ther-
treatment volume. Unfortunately, as with preceding apy plan was not documented at all in the past studies
issues, disadvantages of the existing, largely retro- and was, therefore, unknown. However, Martel et al.
spective and literature apply here as well. Neverthe- (1999) showed that three-dimensional conformal
less, it seems that this issue is focusing on the issue of radiation therapy used to deliver starting doses of
using or omitting elective nodal irradiation, which 69.3–84 Gy to gross tumor volume resulted in 100%
would be a synonym for elective radiation therapy of of the ipsilateral hilum, 59% of the low paratracheal
hilum with or without a part or whole of the medi- region, 57% of the aortopulmonary region, 97% of the
astinum in cases of stage I or a part or whole of the subcarinal region and 57% of the contralateral hilum
mediastinum in stage II non-small-cell lung cancer. receiving C50 Gy. Another report from the same
324 B. Jeremić et al.

institution (Hayman et al. 2001) showed no isolated So far, there is only one prospective randomized
elective nodal failures when three-dimensional con- trial which evaluated the issue of elective nodal
formal radiation therapy was used in patients with irradiation in this setting (Yuan et al. 2007). In this
non-small-cell lung cancer, although there were trial 200 patients were randomized between involved-
3(6%) failures in the lymph nodes outside the plan- field radiation therapy to 68–74 Gy and elective nodal
ning target volume. Rosenzweig et al. (2001) also irradiation to 60–64 Gy. Patients in the involved-field
used similar three-dimensional conformal radiation arm achieved better overall response rate (90 vs. 79%,
therapy (range, 50.4–81 Gy; median, 68.4 Gy) with- p = 0.032) and better 5-year local control rate (51 vs.
out elective nodal radiation therapy to observe 2-year 36%, p = 0.032) than those in the elective nodal
rate of elective nodal control in 88% patients with irradiation arm. Unfortunately, it remained unclear if
tumors locally controlled. With unintentional nodal the poorer outcome from elective nodal irradiation
radiation therapy, a dose of [40 Gy was delivered to was due to the lower radiation therapy dose or the use
ipsilateral superior mediastinum in 34% patients, to of elective nodal irradiation.
the inferior mediastinum in 63% patients and to the When focusing on the issue of elective nodal
subcarinal region in 41% patients. It is, therefore, irradiation one must also consider the incidence of
obvious that not just conventional radiation therapy occult lymph node (hilar and/or mediastinal) metas-
but also three-dimensional conformal radiation ther- tasis. If the initial clinical staging based on comput-
apy (using ‘‘limited’’ radiation therapy fields, e.g., erised tomography scanning is ultimately verified
those covering only macroscopically/radiographically during operation, the incidence of nodal metastases in
visible tumor) frequently result in higher dose to the stage I non-small-cell lung cancer may be as high as
nodal regions that one may initially assume. If one 26% (Glazer et al. 1984; Heavey et al. 1986; Black
intends to document the necessity of elective nodal et al. 1988; Conces et al. 1989), supporting, thus, a
radiation therapy in this setting, a policy of clear consistent finding over the decades that surgical/
documentation of the dose to the regions presumably pathological upstaging is seen in approximately 25%
harboring microscopic spread must be mandatory. of cases of T1N0 and approximately 35% of cases of
Unfortunately, even most recent publications on T2N0 cases (Martini and Beattie 1977; Naruke et al.
the use of three-dimensionally conformal radiation 1988; Ginsberg et al. 1995). Data from surgical
therapy in inoperable non-small-cell lung cancer, studies also showed similar incidence of unsuspected
including more or less cases of early stage non-small- lymph node metastasis when T1 stage was divided by
cell lung cancer, do not document incidental nodal tumor size, 18% in T1a (\2 cm) and 23% in T1b
irradiation, yet claiming that no elective irradiation (2–3 cm) tumors (Koike et al. 1998). They have,
was performed (Belderbos et al. 2003; Bradley et al. therefore, effectively supported previous findings of
2003; Lagerwaard et al. 2002). surgical studies in early stage non-small-cell lung
In some studies institutional policy regarding cancer which clearly showed increasing incidence of
elective nodal irradiation did not change over the time lymphatic invasion/metastasis which occurs with
(Jeremic et al. 1997, 1999), while some (Sandler et al. increasing size of the tumor (\1.0 cm, 1.1–2.0 cm and
1990; Graham et al. 1995) did not provide results [2.0 cm had approximately 0, 17 and 38% of such
according to radiation therapy volume. While Doso- incidence, respectively) (Ishida et al. 1991). When
retz et al. (1992) found no impact of elective nodal immunohistochemical staining was used in patients
irradiation on the treatment outcome, Kupelian et al. with peripheral adenocarcinoma of B2.0 cm, occult
(1996) and Sibley et al. (1998) found better overall nodal (hilar and/or mediastinal) (micro) metastases
survival, disease-specific survival and local control in were detected in 20% of patients (Wu et al. 2001).
patients undergoing elective nodal irradiation, though Additionally, on multivariate analysis, nodal micro-
insignificant, probably due to a small number of metastasis was an independent prognosticator of sur-
events. Morita et al. (1997), however, clearly docu- vival, which was in agreement with previous studies
mented superior complete response rates and overall (Chen et al. 1993; Passlick et al. 1996; Dobashi et al.
survival in patients undergoing elective nodal irradi- 1997; Maruyama et al. 1997). Also, occult nodal
ation and lower distant metastasis rate in patients metastases were significantly more frequent in poorly
undergoing elective nodal irradiation. differentiated tumors, confirming previous findings
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 325

(Takizawa et al. 1998). Although direct comparison advanced disease in 12% of the patients as compared
between surgery and radiation therapy regarding this to chest tomography with false negative findings in 11
issue is not likely to be performed, nevertheless, patients (13%). In a series by Marom et al. (1999)
widely adopted philosophy of the surgical approach (52) nodal staging by computerised tomography,
suggested as preferred/mandatory treatment for T1N0 positron emission tomography and histopathological
patients (i.e., lobectomy) would include systematic analysis disclosed N0 status in 42, 29, and 32 patients,
removal of all hilar and mediastinal lymph node respectively. However, with regard to hilar lymph
content (Ishida et al. 1991; Ginsberg et al. 1995). Its node involvement, both computerised tomography
radiotherapeutic equivalence would be radiation and positron emission tomography were positive in 6
therapy field instituted to treat some, if not all, lymph patients, three of whom were confirmed by patho-
node regions (hilar and/or mediastinal). logical analysis. Hence, the rates of false-negative
On the other side, recent review of the data on the staging by computerised tomography and false posi-
patterns of recurrence after radical radiation therapy tive staging by positron emission tomography were
in early stage non-small-cell lung cancer available in due to differences in mediastinal lymph node assess-
the literature (Jeremic et al. 2002) showed that pre- ment which is of particular importance when assess-
dominant type of failure remains local, being reported ing the role of positron emission tomography in stage
as either isolated or initial in approximately 11–55% I/II lung cancer. These findings are supported by a
cases (ultimate up to 75%). An isolated/initial regio- study by Saunders et al. (1999b) (53) who assessed
nal failure was reported to occur in only 0–7% cases the rate of N2 and N3 mediastinal lymph node
(ultimate up to 15% cases), while the distant metas- involvement by positron emission tomography and
tasis mostly lies between these two (isolated/initial in computerised tomography in a series of 97 patients
3–33% cases and ultimate in up to 36% cases). These under consideration for surgery. True negative find-
findings could support the use of more localized ings were observed in 65 and 62 patients for positron
fields, because it was stressed that the major concern emission tomography and computerised tomography,
should be the gross tumor burden and not a micro- respectively. However, the rate of false negative
scopic one (Williams et al. 2000). However, in the findings differed substantially with 5 patients by
recent study of Yu et al. (2008) in elderly patients positron emission tomography and 12 patients by
([70 years of age), involved field radiation therapy computerised tomography. Thus, positron emission
led to 36.7% elective nodal failures. Importantly, the tomography offers particular advantages of comput-
median time to elective nodal failure was 55 months erised tomography imaging with regard to exclusion
(range, 49–61 months). These figures may, at least of mediastinal lymph node involvement while
partially, explain why radiation therapy-alone studies assessment of hilar nodal disease may be equally
along with short (e.g., 3 years) follow-up are likely to difficult by computerised tomography and positron
underestimate true incidence of isolated nodal failures emission tomography. It is expected that positron
when one uses involved-field radiation therapy in this emission tomography may help better to delineate the
disease. Recent use of positron emission tomography tumor itself, exclude possible areas of regional/distant
scanning in lung cancer has shown that it may be spread, and enable dose-escalation which seems to be
successful not just in detecting subclinical distant possible in the cases with limited lung volume
spread (MacManus et al. 2001) but also in detecting a included in the radiation therapy fields. Finally,
subclinical regional spread. Farrell et al. (Farrell et al. additional advantage of the positron emission
2000) investigated 84 patients without hilar or medi- tomography is that it can be used for the purpose of
astinal lymph node enlargement on computed tomog- treatment planning, owing to increased capability of
raphy. Histopathological N0 disease was confirmed in image fusion (with computerised tomography).
73 patients, 63 of whom had no hilar or mediastinal Radiation targeting with fused FDG-PET and CT
activity on fluorodeoxyglucosae-positron emission images altered the radiation therapy volume in over
tomography scan (86%) while hilar or mediastinal 58% of patients in comparison with CT targeting
lymph node activity and distant metastases were found (Bradley et al. 2004). One of the significant advan-
in 3, 6, and 1 patient, respectively. Thus positron tages of PET is its ability to discriminate tumor tissue
emission tomography rather overestimated more from atelectasis. Decreases in the target volumes in
326 B. Jeremić et al.

patients with atelectasis lead to decreases in normal- with moderately differentiated lesions measuring
tissue toxicity. B1.5 cm in diameter.
Nevertheless, more information on various bio- One of the challenges in radiation oncology is how
logical properties and actual differences between to accurately define microscopic tumor extension and
various subgroups of patients and tumors (e.g., his- clinical target volume for radiation therapy in patients
tology, tumor grade) are needed. They must be with non-small-cell lung cancer. Grills et al. (2007)
gathered before one can embark on investigation of did a clinicopathologic analysis of microscopic
various radiotherapeutic issues in this disease. In one extension in T1N0 lung adenocarcinoma in 35
such attempt Sawyer et al. 1999) used the data patients who underwent surgical resection. The mean
obtained from 346 patients undergoing complete microscopic extension beyond the gross tumor was
resection of early clinical stage non-small-cell lung 7.2 mm and the margin required to cover microscopic
cancer to identify predictors of subclinical nodal extension in 90% of patients was 9.0 mm. Giraud
involvement. Findings of preoperative bronchoscopy, et al. (2000) did a similar analysis in 70 non-small-
tumor size, tumor grade and histology were all com- cell lung cancer surgical resections specimens and
bined to create risk groups for N1/N2/local/regional found that the microscopic extension was different
recurrence; they have found that the risk of subclini- between adenocarcinoma and squamous-cell carci-
cal nodal involvement was at least 15.6% in the best noma. To cover 95% of microscopic extension, a
(low risk) subgroup, while all other patients had margin of 8 and 6 mm must be added for adenocar-
atleast 35% of such a risk. Increasing risk correlated cinoma and squamous-cell carcinoma, respectively.
with increasing size and grade of tumor, accompanied Patients with early stage non-small cell lung cancer
with positive findings of bronchoscopy. In a similar frequently received only fine-needle aspiration biopsy
approach, Suzuki et al. (2001) attempted to determine for pathologic diagnosis. Although fine-needle aspi-
predictors of lymph node and intrapulmonary metas- ration can usually distinguish between small-cell lung
tasis in 389 patients with clinical stage IA non-small- cancer and non-small-cell lung cancer, it is difficult to
cell lung cancer undergoing major lung resection and distinguish between different histologic subtypes of
complete mediastinal lymph node dissection. Of non-small-cell lung cancer (Edelman and Gandara
them, eighty-eight (23%) patients had pathological 2009). As a result, pathology reports sometimes
lymph node involvement or intrapulmonary metasta- describe only ‘‘malignant cells consistent with non-
ses. Significant predictors of local or regional spread small-cell lung cancer’’ and radiation oncologists
included grade of differentiation and pleural come into dilemma how to define microscopic
involvement. When both risk factors were present, extension and what margin to apply.
more than 40% of clinical stage IA non-small-cell Currently, in radiation oncology a treatment plan-
lung cancer patients had pathologic involvement of ning CT with contiguous 3 mm slice thickness is
lymph nodes or intrapulmonary metastases. The same usually required to define gross tumor volume (GTV),
group (Suzuki et al. 1999) previously investigated clinical target volume (CTV) and planning target
clinical predictors of N2 disease in 379 patients with volume (PTV). If elective nodal irradiation is not
clinical N0–N1. There were 68 (17.9%) patients with used, the primary tumor and clinically positive lymph
pathologic N2 stage. Multivariate analysis showed nodes seen on CT ([1 cm in short axis diameter) or
that tumor size, high serum carcinoembryonic antigen pre-treatment PET scan (maximum SUV [2.5) typi-
level and adenocarcinoma histology were significant cally constitute the GTV. The GTV in the lung should
predictors of N2 disease. Finally, Fuwa et al. (2008) be delineated with the ‘‘lung window’’ setting, while
investigated the indication for radiotherapy in 396 mediastinal GTV should be outlined with the
patients with operable peripheral early (T1N0) non- ‘‘mediastinal window’’ setting. Additional 0.5–1.0 cm
small-cell lung cancer by using the data such as age, margin is added to the GTV to account for micro-
gender, Brinkmann’s index, histology, grade of dif- scopic tumor extension and create the CTV. Typi-
ferentiation, tumor diameter and the level of carci- cally, 1.5 cm in the superior-inferior dimensions and
noembryonic antigen as factors involved in lymph at least 1.0 cm in the in the axial plane is added to the
node metastasis. They showed that this risk was low CTV to create the PTV. While CTV margin accounts
in those with well differentiated carcinoma and those for microscopic tumor extension, the PTV margin
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 327

accounts for tumor motion and set-up error. If Kennelly 1980; Sandler et al. 1990; Hayakawa
4DCT scan is available, tumor motion can be seen et al. 1992; Rosenthal et al. 1992; Slotman and
during a respiratory cycle and maximum intensity Karim 1994; Gauden et al. 1995; Morita et al.
projection (MIP) dataset (Underberg et al. 2005) is 1997; Jeremic et al. 1997, 1999; Lagerwaard et al.
used to generate internal target volume (ITV), 2002; Zhao et al. 2007; Pemberton et al. 2009).
which includes tumor and its respiratory motion. An Fang et al. (2006), however, showed that male
additional margin is added to the ITV to create the gender carries independent influence on overall
PTV, although this margin can be significantly survival and adversely influencing it.
reduced if daily image guidance is used. The PTV Majority of the available reports also observed no
is treated with three-dimensional conformal fields influence of the age on treatment outcome (Morrison
shaped to deliver the prescription dose while et al. 1963; Noordijk et al. 1988; Sandler et al. 1990;
restricting the dose to the normal tissues. When a Kaskowitz et al. 1993; Krol et al. 1996; Jeremic et al.
linear accelerator is used, 6–12 MV energy photons 1997, 1999; Hayakawa et al. 1992; Slotman and
should be used for treatment planning, and doses Karim 1994; Slotman et al. 1996; Gauden et al. 1995;
should be calculated with heterogeneity corrections Rosenthal et al. 1992; Zhao et al. 2007; Pemberton
that take into account the density differences within et al. 2009). This was also shown in the study of
the irradiated volume. Campeau et al. (2009) on the use of radiation therapy
Owing to somewhat conflicting results, identified alone and radiation therapy and concurrent chemo-
when discussing this issue (Jeremic 2004, 2007; therapy. However, Morita et al. (1997) found detri-
Belderbos et al. 2008, 2009) no reliable recom- mental effect of age of [80 years on overall survival,
mendations can be made concerning elective nodal though not providing data on other endpoints such as
irradiation, although prevailing opinion in the year cause-specific survival. Similarly, Sibley et al. (1998),
2011 is to omit elective nodal irradiation. However, however, used both uni- and multivariate analysis to
there seems to be a subgroup of patients with show that younger age positively correlated with
increased risk of developing nodal metastasis, iden- overall survival and cause-specific survival. Both
tification of which must be one of the priorities of studies, however, provide no explanation at all or
research in this field. Contrary to that, it is reason- perhaps a hypothesis for such finding. The same
able to assume that at least for small peripheral, low- remains true for the report of Lagerwaard et al. (2002)
grade tumors it would be the best to administer who found detrimental effect of increasing age
involved-field radiation therapy (omitting elective (patients were grouped as having age \70, 70–75, and
nodal irradiation), due to lowest incidence of occult [75 years) on overall survival, but not on cause-
nodal metastasis. Ultimately, however, more infor- specific survival or local and distant tumor control.
mation regarding biology of these tumors is needed Similar was observed by Firat et al. (2002) in a uni-
because identification of potential factors contribut- variate analysis, though not confirmed by the multi-
ing to higher incidence of subclinical regional lymph variate analysis. Fang et al. (2006) showed that age
node metastasis would help to optimize radiation C70 years was an independent prognosticator of
therapy fields and enable successful dose-escalation inferior survival. Recent analyses focusing on elderly
at the primary tumor level. with early stage non-small-cell lung cancer (Furuta
et al. 1996; Hayakawa et al. 2001; Gauden and
Tripcony 2001) showed similar outcome for this
6 Prognostic Factors patient population when treated with radiation therapy
alone, which may go as high as 36% when 5-year
In addition to radiation therapy-related factors, there cause-specific survival was used as an endpoint
were also attempts to investigate the influence of (Furuta et al. 1996).
various pre-treatment, both patient- and tumor- Influence of the performance status on treatment
related, prognostic factors using either overall outcome is still controversial. Both Dosoretz et al.
survival or cause-specific survival/disease-specific (1992), Slotman and Karim (1994), Kaskowitz et al.
survival as endpoints. Gender seems not to play a (1993) (23), Gauden et al. (1995) and Pemberton
major role in the outcome of patients (Coy and et al. (2009) found no influence of performance status
328 B. Jeremić et al.

on either overall survival or disease-specific survival,


while Rosenthal et al. (1992), Hayakawa et al. (1992), 7 Toxicity
Kupelian et al. (1996) (14), Jeremic et al. (1997,
1999) as well as Hsie et al. (2007) did note its effect Although treatment-related deaths have already been
on either overall survival and/or disease-specific sur- documented in as early as the very first report
vival/relapse-free survival as well. In the study of (Morrison et al. 1963) after 45 Gy given in 20 daily
Lagerwaard et al. (2002) the World Health Organi- fractions over 4 weeks, this issue has not been sys-
zation performance status score was an independent tematically addressed over the years. Frequently
prognosticator of OS, and the same was observed in authors did not provide information on toxicity at all
the study of Firat et al. (2002) using the Karnofsky (Coy and Kennelly 1980; Cooper et al. 1985; Zhang
performance status score in a univariate analysis. et al. 1989; Rosenthal et al. 1992; Krol et al. 1996),
Likewise, conflicting results are seen with weight loss while others only mentioned either absence or rarity
(Fang et al. 2006; Pemberton et al. 2009; Campeau of, mostly ‘‘serious’’, toxicity (Noordijk et al. 1988;
et al. 2009). Sandler et al. 1990; Dosoretz et al. 1992; Gauden
Investigating influence of histology on treatment et al. 1995; Slotman et al. 1996; Morita et al. 1997;
outcome, only Sibley et al. (1998) found an Hayakawa et al. 1999). When data were provided
improvement in cause-specific survival for squamous without specifying the toxicity criteria, there was
histology, while Gauden et al. (1995) observed so for usually no report on high-grade (C3) toxicity. Mild to
the mixed (adenocarcinoma/squamous-cell carci- moderate (corresponding to grades 1 and 2) esopha-
noma) histology using both overall survival and gitis was usually seen in up to two-thirds of patients,
relapse-free survival as endpoints. Lagerwaard et al. while mild to moderate pneumonitis was usually seen
(2002) observed an independent and favorable influ- in approximately 20% of patients. This was observed
ence of unknown histology (versus squamous-cell regardless of tumor/dose fractionation pattern or
histology and non-squamous-cell histology) on over- whether elective nodal radiation therapy was used or
all survival. All other studies observed no such effect not. However, Hayakawa et al. (1992) described four
(Sandler et al. 1990; Dosoretz et al. 1992; Hayakawa out of 13 (31%) patients dying of pulmonary insuffi-
et al. 1992; Rosenthal et al. 1992; Dosoretz et al. ciency due to bronchial stenotic changes after
1993; Slotman and Karim 1994; Slotman et al. 1996; receiving [80 Gy in 2 Gy daily fractions at the
Jeremic et al. 1997; Firat et al. 2002; Pemberton proximal bronchi.
et al. 2009), including radiochemotherapy studies There were only several studies that reported on
(Campeau et al. 2009). Only Hayakawa et al. (1992) toxicity using grading systems. Graham et al. (1995)
observed better outcome for tumors located in the reported on mild to moderate acute toxicity in 103
upper lobes or the superior segment of the lower patients with early stage non-small-cell lung cancer
lobes, while all other studies excluded its possible treated with 18–60 Gy (median primary dose, 60 Gy
effect when comparing central versus peripheral in 30 fractions), of whom 80% received elective
location (Ono et al. 1991; Slotman and Karim 1994; nodal radiation therapy. One patient developed
Slotman et al. 1996; Jeremic et al. 1997; Cheung et al. European Organization for the research and treat-
2000; Lagerwaard et al. 2002). ment of Cancer/Radiation Therapy Oncology Group
In addition to various clinical prognostic factors, a (Cox et al. 1995) grade 3 pneumonitis and there
number of biological and molecular characteristics of were also only three cases of late grade 2 lung tox-
lung cancer may influence treatment outcome. While icity. Jeremic et al. (1997) treated 49 patients with
they have been investigated in surgical patients stage I non-small-cell lung cancer with hyperfrac-
(Slebos et al. 1990; Tateishi et al. 1991; Fontanini tionated radiation therapy doses of 69.6 Gy via
et al. 1992; Pastorino et al. 1997), these information’s 1.2 Gy b.i.d. fractionation to show only 2 (6%) grade
are basically lacking in medically inoperable early 3 acute toxicities (bronchopulmonary and esophageal)
stage non-small-cell lung cancer patients treated and 3 (9%) grade 3 late toxicities (bronchopulmonary,
with radiation therapy either alone or given with esophageal and osseous), although the ipsilateral
chemotherapy. hilum was electively treated to a 50.40 Gy dose with
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 329

the same fractionation. Jeremic et al. (1999) again Major problem with the data from the literature is a
used Radiation Therapy Oncology group (Cox et al. great variety of both pre-treatment and radiation ther-
1995) toxicity criteria to report on the same hyper- apy-related factors, such as the total dose, fractionation
fractionated radiation therapy regimen (69.6 Gy using or treatment fields, not just between the institutions, but
1.2 Gy b.i.d. fractionation) in 67 stage II non-small- intrainstitutionally, too. These have greatly obscured
cell lung cancer patients. Although elective medias- the overall picture and prohibited us from having firm
tinal irradiation was used in all cases, there were only conclusions. While it is a well-established premise that
2 bronchopulmonary and two esophageal acute grade higher total dose, higher dose per fraction, and larger
3 toxicities (total n = 4.6%) and only one broncho- volume of the lung irradiated should lead to more
pulmonary and two esophageal late grade 3 toxici- toxicity (Moss et al. 1960; Holsti and Vuorinen 1967;
ties (total n = 3.4%). Sibley et al. (1998) observed Rubin and Casarett 1968; Mah et al. 1987; McDonald
2 (1.5%) grade C3 complications, one being fatal et al. 1995), both acute and late, it is unknown to what
pneumonitis 2 months after the completion of extent other, radiation therapy-unrelated factors such as
66 Gy in 2 Gy fractions, the other being severe pre-existing comorbidity, infections, or simply natural
oxygen-dependent pneumonitis unresponsive to ste- processes such as sclerosis present in elderly, may add
roids after 64 Gy in 2 Gy fractions. Both patients to the occurrence of toxicity (Rubin and Casarett 1968;
had their mediastinum region encompassed. How- Braun et al. 1975; Prasad 1978; Garipagaoglu et al.
ever, no information on grading system used in that 1999). Some, however, have shown that concomitant
study was provided. Finally, Lagerwaard et al. chronic obstructive pulmonary disease did not increase
(2002) observed grade 1–2 esophagitis according to the risk of radiation pneumonitis (Prasad 1978).
the Radiation Therapy Oncology Group in 16% Acute high-grade toxicity may also be interesting
patients with no symptoms consistent with late as a contributing (causal) factor leading to more
esophageal toxicity. Grade 2 or higher on South- treatment interruptions which, on the other side, may
west Oncology Group scale was observed in 6% adversely influence treatment outcome (Cox et al.
patients. In the latter group of reports (Jeremic 1993; Chen et al. 2000; Jeremic et al. 2003b). In the
et al. 1997, 1999; Graham et al. 1995; Sibley et al. first-ever analysis devoted to this issue exclusively in
1998), high-grade (C3) acute esophagitis and early stage non-small-cell lung cancer (Jeremic et al.
pneumonitis were documented in up to 3% of cases, 2003b), of a total of 116 patients treated with total
and the same applies to the high-grade late toxicity, tumor doses of 69.6, 1.2 Gy b.i.d. fractionation, 44
with no apparent differences between various radi- patients refused surgery while 72 patients were
ation therapy regimens, although Lagerwaard et al. medically inoperable due to existing co-morbid states.
(2002) used multivariate analysis to document det- Medically inoperable patients had worse KPS
rimental effect of radiation dose of 70 Gy or more (p = 0.0059) and more pronounced weight loss
on the incidence of acute esophagitis. (p = 0.0005). Among them, 12 patients experienced
In none of these series it was not specifically high-grade toxicity and 11 out of these 12 patients
reported that these toxic events have happened in were with either acute (n = 6) or ‘‘consequential’’
elderly patients. When, however, study population late (n = 5) high-grade toxicity that requested inter-
was completely confined to elderly with early stage ruption in the hyperfractionated radiation therapy
non-small-cell lung cancer, no significant radiation course (range, 12–25 days; median, 17 days). Patients
therapy-related complications were found and inci- who refused surgery achieved superior survival
dence of both acute and late high-grade (3 and 4) when compared to medically inoperable patients
toxicity was similar among all age groups (Gauden (p = 0.0041), as well as superior local recurrence-
et al. 1995; Gauden and Tripcony 2001). Even when free survival (p = 0.011), but not different distant
radiation therapy-related deaths occurred and were metastasis-free survival (p = 0.14). Cause-specific
reported on (Hayakawa et al. 2001), again, there was survival also favored patients who refused surgery
no difference between elderly (5%) treated with (p = 0.004). Multivariate analysis showed indepen-
highest dose levels (80 Gy) and their non-elderly dent influence as the reason for not undergoing
counterparts (4%) treated the same way. surgery on overall survival (p = 0.035), but not on
330 B. Jeremić et al.

local recurrence-free survival (p = 0.084) or cause- step in quantitative assessment of competitive treat-
specific survival (p = 0.068). Patients who refused ment plans and a screening tool to select ‘‘the best’’
surgery did not experience high-grade toxicity (0/44), available plan (Drzymala et al. 1991), usually coupled
whereas 11 of 72 patients with medical inoperability with other quantitative indices such as normal-tissue
and co-morbid states experienced high-grade toxicity complication probability and tumor control probabil-
and had treatment interruptions to manage toxicity ity (Kutcher and Burman 1987; Lyman and Wolbarst
(p = 0.0064). Patients without treatment interruptions 1987; Burman et al. 1991). They may enable an
had significantly better overall survival (p = 0.00000), increase in the dose delivered to tumor, necessary for
local recurrence-free survival (p = 0.00000), and better tumor control (Armstrong et al. 1993;
cause-specific survival (p = 0.00000) than those with Robertson et al. 1997). they can also give useful data
treatment interruptions. When corrected for treatment for characterization of the dose–volume relationship
interruptions, the reason for not undergoing surgery and the development of pneumonitis (Martel et al.
independently influenced overall survival (p = 0.040), 1994; Oetzel et al. 1995; Kwa et al. 1998; Graham
but not local recurrence-free survival (p = 0.092) or et al. 1999) and reduced dose to not just lung (Graham
cause-specific survival (p = 0.068). In contrast to this, et al. 1994), but other critical normal tissues as well
treatment interruption was independent prognosticator (Maguire et al. 1999; Bahri et al. 1999). Multivariate
of all three endpoints used (p = 0.00031, p = 0.0075 analysis by Graham et al. (1999) revealed that the
and p = 0.00033, respectively). When 11 patients with total lung volume receiving 20 Gy (V20) to be the
treatment interruptions were excluded, the reason for single independent predictor of pneumonitis. In this
not undergoing surgery still affected overall survival study with three-dimensional treatment planning,
(p = 0.037) and cause-specific survival (p = 0.039) actuarial incidence of [Grade 2 pneumonitis by
but not local recurrence-free survival (p = 0.11). 24 months was 7% for V20 between 22 and 31%, and
Multivariate analyses using overall survival, cause- 13% for V20 between 32 and 40% (Fig. 3). Nowa-
specific survival and local recurrence-free survival days, when intensity modulated radiation therapy is
showed that the reason for not undergoing surgery used; the question remains whether total lung V20 is
affected overall survival (p = 0.0436), but neither enough to predict the incidence of radiation pneu-
cause-specific survival (p = 0.083) nor local recur- monitis. Yom et al. (2007) was among the first to
rence-free survival (p = 0.080). Late high-grade tox- provide clinical data regarding the rate of high-grade
icity also becomes interesting from the standpoint of radiation pneumonitis in patients treated with inten-
prolonged survival of these patients. Prolonged follow- sity modulated radiation therapy. The 12-month
up is, therefore, necessary. It may also be advantageous incidence of radiation pneumonitis in patients with
in terms of detecting second cancers, both lung and lung volume receiving 5 Gy (V5) \70% was 2%,
non-lung, that occur in long-term survivors after the while for those with V5 [70%, the rate was 21%
first radiation therapy (Jeremic et al. 2003a). If diag- (Yom et al. 2007). When 60–70 Gy is delivered in
nosed at an early stage, these patients may experience 1.8–2.0 Gy fractions utilizing three-dimensional
similar outcome as with the first radiation therapy. conformal radiation therapy planning, it is prudent to
Reporting of toxicity poses an additional problem, keep total lung V20 \35%. With intensity modulated
because only rarely scoring systems have been used. radiation therapy planning low dose spillage is higher
Additionally, such reporting was almost always done and it is reasonable to keep total lung V5 \70% as
on an actual (crude) basis, and not on the actuarial suggested by Yom et al. (2007). There is, however, a
one. While the former method may be acceptable, considerable variation in total lung volume definition.
although not preferable, for acute toxicity, it should While some radiation oncologists define the total lung
be strongly discouraged as totally inappropriate for volume as the total lung volume minus the gross
late toxicity. tumor volume, others define this volume as the total
With the wide introduction of computerised three- lung volume minus the clinical target volume, which
dimensional treatment planning in recent years, it is includes gross tumor volume with an expansion for
now widely possible to tailor the dose to tumor and microscopic extension. Since the majority of papers
spare surrounding healthy tissues. In particular, the reporting the incidence of radiation pneumonitis
use of dose–volume histograms enabled a preliminary are retrospective with different total lung volume
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 331

Fig. 3 Dose volume histogram for the treatment plan shown in receiving 20 Gy (V20) was 14.6%. (The clinical target volume
Fig. 1. Note that 95% of the planning target volume (PTV) was subtracted from the total lung volume for total lung V20
receives the prescription dose (70 Gy). Total lung volume calculation.)

definitions, it can be sometimes difficult to compare survival as well as toxicity by weighting the time
data from two different institutions. spent with a specific toxicity as well as local or distant
tumor progression. Each of the number of toxicities
was weighted with increasing severity as the toxicity
8 Quality of Life increased in grade. Nine hundred seventy-nine
patients with stage II–IIIB (vast majority of stage III;
The quality of life in patients treated with radiation no stage I) inoperable non-small-cell lung cancer
therapy becomes increasingly important issue in lung were enrolled on six prospective phase II and III
cancer, but no clear data exist in early non-small-cell studies that ranged from standard radiation therapy
lung cancer treated by radiation therapy alone. To (60 Gy), hyperfractionated radiation therapy
address this issue, Movsas et al. (1999) used a quality- (69.6 Gy), induction chemotherapy followed by
adjusted survival time model which takes into account standard radiation therapy, induction chemotherapy
332 B. Jeremić et al.

and concurrent radiochemotherapy and concurrent metastasis, including those originating from primary
chemotherapy and hyperfractionated radiation ther- lung cancer in mid-eighties of the last century (Sturm
apy. Patients \60 years old had improved survival et al. 1987; Flickinger et al. 1994; Alexander et al.
with more aggressive therapy (with chemotherapy 1995). It had also been shown that stereotactic frac-
added to radiation therapy), while those[70 years old tionated radiation therapy is an effective treatment
achieved the best quality-adjusted survival time with approach for both malignant and nonmalignant neo-
standard radiation therapy alone. The same authors plasms because it combines the accurate focal dose
used the Quality-adjusted time without symptoms delivery of stereotactic radiosurgery with the biolog-
(Q-Twist) in the same group of patients enrolled ical advantages of fractionated radiation therapy
during Radiation Therapy Oncology Group studies in (Dunbar et al. 1994; Kooy et al. 1994; Varlotto et al.
locally advanced non-small-cell lung cancer, a 1996). It was also indicated that stereotactic frac-
minority of whom were stage II (Movsas et al. 2000). tionated radiation therapy can be advantageous over
A quality-adjusted survival analysis subtracted from stereotactic radiosurgery in tumors [3 cm or those
survival time spent with toxicity and/or relapse. located in the vicinity of critical organs (Dunbar et al.
While an overall benefit in Q-Twist was seen with the 1994; Varlotto et al. 1996). This experience led to
addition of chemotherapy to radiation therapy, the application of stereotactic techniques in numerous
advantage of more aggressive therapy was limited to extracranial tumor sites, including that of lung
patients \70 years old. In patients [70 years, no (Blomgren et al. 1995; Uematsu et al. 1998; Uematsu
radiation therapy/chemotherapy regimen had a supe- et al. 2001; Fukumoto et al. 2002; Nagata et al. 2002;
rior Q-Twist than radiation therapy alone. None of Hara et al. 2002; Hof et al. 2003; Onimaru et al. 2003;
these analyses provided separate analysis for patients Whyte et al. 2003; Lee et al. 2003; Timmerman et al.
with early stage non-small-cell lung cancer. 2003). While initial studies included patients with
A study by Langendijk et al. (2000) on the pre- lung metastasis and those with early stage non-small-
treatment quality of life in inoperable non-small-cell cell lung cancer, more recent reports concentrated
lung cancer (stage I, 21%; stage II, 1%) disclosed that exclusively on the latter. Separate chapter focusses on
World Health Organization performance status, the use of stereotactic radiation therapy in early non
weight loss, and age were all significantly associated small cell lung cancer. Breiefly, though, suffice to say
with quality of life. Among the different respiratory that initial results were indeed impressive. Local
symptoms assessed by the European Organization for tumor control was obtained in at least 85% of
the research and treatment of cancer quality of life patients, while 2–3 year survivals went up to 60–70%,
questionnaire-C30 score, dyspnoea was the only item all accompanied with very low toxicity. Recently
that significantly correlated with global quality of life. published prospective phase II data in medically
Furthermore, changes of dyspnoea subsequent to inoperable patients with early stage non-small-cell
treatment were significantly associated with global lung cancer demonstrated 3-year primary local tumor
quality of life as well. Unfortunately, neither analysis control [90% and 3-year overall survival of 56–60%
of treatment-related toxicity and quality of life was (Baumann et al. 2009; Timmerman et al. 2010).
included in this study nor there was a separate anal- While detailed overview of the technological and
ysis relating to early stage non-small-cell lung cancer. biological principles would follow in a latter chapter,
suffice to say that these results await longer followup
and possible comparison with sublobar resection in
9 Novel Approaches patients who cannot tolerate lobectomy. Prospective
randomized trials comparing stereotactic radiation
9.1 Stereotactic Radiation Therapy therapy with conventionally fractionated radiation
therapy are also warranted since the majority of
Stereotactic radiation therapy is an extremely hypo- published data with conventional fractionation come
fractionated form of radiation therapy for early stage from the era of 2D planning. With no doubt, com-
non-small-cell lung cancer. Historically, high preci- parison of this data with modern data from the 3D era
sion radiation therapy in a form of stereotactic radi- and stereotactic body radiation therapy is an unfair
osurgery was successfully introduced in cases of brain comparison.
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 333

9.2 Accelerated Hypofractionated patients, mostly medically inoperable with periph-


Radiation Therapy erally located T1-3N0 non-small-cell lung cancer.
A dose of 48–60 fractions was delivered in 4 Gy
Accelerated hypofractionated radiation therapy is a fractions with most patients receiving 48–52 Gy.
moderately hypofractionated form of radiation ther- Gross tumor volume was treated with 1.0–1.5 cm
apy for early stage non-small-cell lung cancer. While margin without elective nodal irradiation. The 2- and
stereotactic body radiation therapy typically employs 5-year local control rates were 76.2 and 70.1%,
C8 Gy, accelerated hypofractionated radiation ther- respectively. The 2- and 5-year overall survival rates
apy employs lower doses of radiation therapy, usually 51 and 23.3%, respectively, while median overall
3.5–4.0 Gy fraction to a total cumulative dose of survival was 26.5 months. Based on this experience,
48–60 Gy. Although high doses with conventionally the National Cancer Institute of Canada opened a
fractionated 1.8–2.0 Gy radiation therapy were fea- Phase II trial of 3D conformal radiation therapy to
sible when the volume of irradiated lung was limited, deliver 60 Gy in 4 Gy fractions for medically inop-
overall treatment time was extended to more than erable patients with T1-3N0 non-small-cell lung
7 weeks. For many medically inoperable patients with cancer (NCIC BR.25) The study was conducted
early stage non-small-cell lung cancer even a 7-week between 2006 and 2008 and the results from
course of radiation therapy can be difficult since the this prospective, multi-institutional trial are pending
majority of patients have serious comorbidites. Thus, (Soliman et al. 2011).
accelerated hypofractionated radiation therapy allows Bogart et al. (2010) in the United States reported a
a shorter treatment course without typical require- phase I dose-escalation study of accelerated hypo-
ments for stereotactic body radiation therapy such as fractionated radiation therapy in 39 medically inop-
accurate patient immobilization, precise accounting erable patients with T1-2N0 non-small-cell lung
for tumor motion and, image guidance. cancer (CALBG 39904). A dose of 70 Gy was
In Slotman et al. (1996) reported their experience delivered while the daily fraction size was escalated
in 31 patients with T1-2N0 non-small-cell lung can- from 2.41 Gy in 29 fractions to 4.11 Gy in 17 frac-
cer. A dose of 48 Gy in 4 Gy fractions was delivered tions. Without elective nodal irradiation, gross tumor
with five fractions per week over a period of two and volume with at least 1.5 cm margin was treated with
a half weeks. Gross tumor volume was treated with a 3D conformal radiation therapy and heterogeneity
margin of at least 1.5 cm for the first 40 Gy and a correction. The major response rate was 77% (31%
margin of at least 0.5 cm for the boost phase with no complete response, 46% partial response) and 23% of
elective nodal irradiation. The majority of patients patients had stable disease. After a median follow-up
were medically inoperable (87%) and none of the time of 53 months, median overall survival was
patients underwent a surgical staging procedure. The 38.5 months. Treatment was well tolerated without
mean tumor diameter was 3.2 cm and most tumors grade 4 or higher toxicity.
were peripherally located. A recurrence was seen in Obviously, the intent of accelerated hypofraction-
19% of patients, 3-year overall survival was 42% and ated approach is not to develop a new type of ste-
median overall survival was 33 months. Faria et al. reotactic radiation therapy, but rather to find an
(2006) in Canada published their results in 32 patients alternative to extreme hypofractionation with the
with early stage non-small-cell lung cancer. Most stereotactic approach. Accelerated hypofractionated
patients had T1-2N0 disease and although T1-2N1 radiation therapy does not require a stereotactic type
patients were eligible, 94% of patients had N0 disease of immobilization, precise accounting for tumor
by CT staging. With 3D planning, a dose of 52.5 Gy motion and prolonged time on treatment couch nec-
in 3.5 Gy fractions was delivered with five fractions essary to deliver each fraction of radiation therapy.
per week over a period of 3 weeks. Gross tumor For patients who cannot tolerate stereotactic radiation
volume with a 1.0–1.5 cm margin was treated without therapy immobilization or treatment process, accel-
elective nodal irradiation. The 2-year local relapse- erated hypofractionated radiation therapy can be an
free survival was 76%, 2-year overall survival 56% alternative approach. This moderately hypofraction-
and median overall survival 29 months Another ated regimen has a potential to become an alternative
Canadian study by Soliman et al. (2011) included 118 to stereotactic radiation therapy, particularly in small
334 B. Jeremić et al.

community radiation oncology practices if further was observed. Late high-grade toxicity was rarely
prospective clinical trials confirm its efficacy. observed (total 10%). The patient population in this
study was very favorable. The majority of patients
were in a good Karnofsky performance Status score,
9.3 Accelerated Hyperfractionated none had weight loss of [5 and 70% of patients
Radiation Therapy and Concurrent enrolled into this study actually refused surgery.
Radiation Therapy Campeau et al. (2009) recently reported on 73 patients,
with Chemotherapy who were treated either with concurrent radiation
therapy and chemotherapy (n = 39) or radiation
Evidence also slowly emerged for the use of concur- therapy alone using total dose of either 60 Gy
rent radiation therapy and chemotherapy, although it (n = 23) or 50–55 Gy (n = 11). Intention-to-treat
has been done only rarely. In an Australian study (Ball analysis showed that the 2-year survival for concurrent
et al. 2001), 24 patients randomized to concurrent regimen was 57% and that for radiation therapy alone
carboplatin versus conventionally fractionated radical regimens combined was 33%, clearly favoring con-
RT (60 Gy) alone achieved an MST of 41.6 months current regimen. Also, the 2-year local progression-
versus 19.5 months and an estimated 2-year survival free survival was 66 and 55% for the combined and
rate of 77 versus 27% (p = 0.042), although stage was radiation therapy group alone, respectively.
not an independent prognosticator in that study
involving mostly Stage III non-small-cell lung cancer
patients. A recent subset analysis (Bentzen et al. 2000) 10 Conclusions
of 169 patients with stage I–IIA non-small-cell lung
cancer initially enrolled in the continuous hyperfrac- A proportion of early stage non-small-cell lung cancer
tionated accelerated radiotherapy (CHART) study undergoes radiation therapy alone due to several
(Saunders et al. 1999a) showed a benefit of 13% at reasons. Although this patient population must be
2 years (37 vs. 24%) and 6% at 4 years (18 vs. 12%) considered unfavorable, radiation therapy alone
for CHART (54 Gy) over conventionally fractionated remains standard treatment option in these patients
radical radiation therapy (60 Gy). In a recent pro- which are frequently named as technically operable
spective phase II trial, Jeremic et al. (2005) used but medically inoperable. Although survival figures
concurrent hyperfractionated radiotherapy and low- are still lower than those obtainable with surgical
dose daily chemotherapy consisting of carboplatin and candidates, even when clinically staged, with con-
paclitaxel in a phase II study. Fifty-six patients started ventional high-dose radiation therapy the median
their treatment on day 1 with 30 mg/m2 of paclitaxel. survival times of up to 35 months and 5-year survival
Hyperfractionated radiotherapy using 1.3 Gy b.i.d. to of up to 35% have been obtained. These figures are
a total dose of 67.6 Gy and concurrent low-dose daily even better when cause-specific survival is used.
carboplatin 25 mg/m2 and paclitaxel 10 mg/m2, both Various radiation therapy characteristics are exam-
given daily (but excluding weekend days) during the ined showing that there seems to be a favorable effect
radiation therapy course, starting from the second day. of high doses on outcome, as well as it seems to be
The median survival time was 35 months, and 3- and favorable effects of smaller size/lower stage. While
5-year survival rates were 50 and 36%, respectively. there is no general agreement on the use of elective
The median time to local progression has not been nodal irradiation, some tumors (e.g., small, peripheral
achieved, but 3- and 5-year local progression-free lesions) seem as the most suitable for limited radia-
survival rates were 56 and 54%, respectively. The tion therapy. Unfortunately, discrepancies between
median time to distant metastasis has not been surgical and radiotherapeutic series regarding the
achieved, but 3- and 5-year distant metastasis-free staging procedures, treatment procedures in this dis-
survival rates were 61 and 61%, respectively. The ease as well as the documentation of the pattern of
median and 5-year cause-specific survivals were failure make any conclusion unreliable. They, how-
39 months and 43%, respectively. Acute high-grade ever, call for more cooperation between technology
(C3) toxicity was hematological (22%), esophageal and biology in order to more selectively apply one or
(7%), or bronchopulmonary (7%). No grade 5 toxicity another approach. Suggesting that limited field
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 335

radiation therapy must be used for the sake of dose versus 19.5 months and an estimated 2-year survival
escalation (leading to better tumor control and/or less of 77 versus 27%, p = 0.042), although stage was not
toxicity) would inevitably lead to misuse of these an independent prognosticator in that study involving
technologies and false interpretation of the results. majority of stage III non-small-cell lung cancer
Pattern of failure after radiation therapy alone clearly patients. In a recent subset analysis (Bentzen et al.
identified local component as predominant, while 2001) of 169 patients with stage I–IIA non-small-cell
observed rare isolated nodal relapses are in contrast lung cancer initially enrolled in the continuous
with surgical findings in the same disease. Of a hyperfractionated accelerated radiotherapy study
number of pre-treatment patients and tumor charac- (Saunders et al. 1999a) there was a benefit of 13% at
teristics occasionally examined gender and age 2 years (37 vs. 24%) and 6% at 4 years (18 vs. 12%),
probably do not influence survival. Performance sta- for continuous hyperfractionated accelerated radiation
tus and weight loss may exert its influence on sur- therapy (54 Gy) over conventionally fractionated
vival, but possible effects of tumor location and radical radiation therapy (60 Gy), respectively. It
histology remain controversial. Reported toxicity of showed again that community of radiation oncologists
radiation therapy is confined to mild to moderate dealing with lung cancer must use prospective ran-
bronchopulmonary and esophageal toxicity. Although domized trials to ask simple and meaningful questions
it is reported as rare event, except in cases with very and to obtain answers which may instantly be used in
high doses when given after conventional planning, the clinic, a task having a major importance in this
it’s reporting needs to be substantially improved and disease.
systematically addressed. Quality of life is an issue
completely underrepresented and needs to be focused
upon, especially with expected increase in the long- References
term survivors with the use of sophisticated tools for
treatment planning and delivery which will enable Alexander E 3rd, Moriarty TM, Davis RB, Wen PY, Fine HA,
further dose escalation in this disease. Black PM, Kooy HM, Loeffler JS (1995) Stereotactic
Some, if not all, of the issues discussed above radiosurgery for the definitive, noninvasive treatment of
brain metastases. J Natl Cancer Inst 87:34–40
could have been settled in case of existing prospective Armstrong JG, Burman C, Leibel S, Fontenla D, Kutcher G,
randomized studies. Unfortunately, they are lacking, Zelefsky M, Fuks Z (1993) Three-dimensional conformal
although patients with early stage non-small-cell lung radiation therapy may improve the therapeutic ratio of high
cancer were sometimes included in prospective dose radiation therapy for lung cancer. Int J Radiat Oncol
Biol Phys 26:685–689
studies evaluating the effect of various altered frac- Au J, El-Oakley R, Cameron EW (1994) Pneumonectomy for
tionated regimens, alone or with concurrent chemo- bronchoigenic carcinoma in the elderly. Eur J Cardiothorac
therapy, mostly, however, without specifying its Surg 8:247–250
outcome. Pure hyperfractionated radiation therapy Bahri S, Flickinger JC, Kalend AM, Deutsch M, Belani CP,
Sciurba FC, Luketich JD, Greenberger JS (1999) Results of
alone or with either induction or concurrent chemo- multifield confromal radiation therapy of nonsmall-cell lung
therapy (Cox et al. 1990; Sause et al. 1995; Lee et al. carcinoma using multileaf collimation beams. Radiat Oncol
1996; Komaki et al. 1997), accelerated radiation Investig 7:297–308
therapy via concomitant boost (Byhardt et al. 1993) Ball D, Bishop J, Smith J, O’Brien P, Davis S, Ryan G, Olver I,
Toner G, Walker Q, Joseph D (1999) A randomised phase
were used, but without specifying the outcome in this III study of accelerated or standard fraction radiotherapy
patient population. When accelerated hyperfraction- with or without concurrent carboplatin in inoperable non-
ated radiation therapy using concomitant boost was small cell lung cancer: final report of an Australian multi-
used with doses as high as 73.6–80 Gy, the median centre trial. Radiother Oncol 52:129–136
Ball DL, Peters LJ, Smith J (2001) Untitled (letter to the
survival time for patients with stage I/II was editor). Radiother Oncol 58:89–90
34 months and the median local progression-free Basaki K, Abe Y, Aokli M (2006) Prognostic factors for
survival was 23 months (Maguire et al. 2001). In an survival in stage III non-small-cell lung cancer treatyed with
Australian study (Ball et al. 1999), patients random- definitive radiation therapy. Impact of tumor volume. Int J
Radiat Oncol Biol Phys 64:449–454
ized to concurrent carboplatin versus conventionally Baumann P, Nyman J, Hoyer M, Wennberg B, Gagliardi G,
fractionated radical radiation therapy (60 Gy) alone Lax I, Drugge N, Ekberg L, Friesland S, Johansson KA,
achieved the median survival time of 41.6 months Lund JA, Morhed E, Nilsson K, Levin N, Paludan M,
336 B. Jeremić et al.

Sederholm C, Traberg A, Wittgren L, Lewensohn R (2009) Burman C, Kutcher GJ, Emami B, Goitein M (1991) Fitting of
Outcome in a prospective phase II trial of medically normal tissue tolerance data to analytic function. Int J
inoperable stage I non-small-cell lung cancer patients Radiat Oncol Biol Phys 21:123–135
treated with stereotactic body radiotherapy. J Clin Oncol Byhardt RW, Pajak TF, Emami B, Herskovic A, Doggett RS,
27:3290–3296 Olsen LA (1993) A phase I/II study to evaluate accelerated
Belderbos JSA, De Jaeger K, Heemsbergen WD, Seppenwoolde fractionation via concomitant boost for squamous, adeno,
Y, Baas P, Boersma LJ, Lebesque JV (2003) First results of a and large cell carcinoma of the lung: report of radiation
phase I/II dose escalation trial in non-small cell lung cancer therapy oncology group 84-07. Int J Radiat Oncol Biol Phys
using three-dimensional conformal radiotherapy. Radiother 26:459–468
Oncol 66:119–126 Campeau M-P, Herschtal A, Wheeler G, MacManus MM,
Belderbos JS, Kepka L, Spring Kong FM, Martel MK, Videtic Wirth A, Michael M, Hogg A, Drummond E, Ball D (2009)
GM, Jeremic B (2008) Report from the international atomic Local control and survival following concomitant chemo-
energy agency (IAEA) consultants’ meeting on elective radiotherapy in inoperable stage I non-small-cell lung
nodal irradiation in lung cancer: non-small-cell lung cancer cancer. Int J Radiat Oncol Biol Phys 74:1371–1375
(NSCLC). Int J Radiat Oncol Biol Phys 72:335–342 Cerfolio RJ, Bryant AS, Eloubeidi MA (2006) Routine
Belderbos JS, Kepka L, Kong FM, Martel MK, Videtic GM, mediastinoscopy and esophageal ultrasound fine-needle
Jeremic B (2009) Elective nodal irradiation (ENI) in locally aspiration in patients with non-small cell lung cancer who
advanced non-small-cell lung cancer (NSCLC): evidence are clinically N2 negative: a prospective study. Chest
versus opinion? Int J Radiat Oncol Biol Phys 74:322 130:1791–1795
Bentzen SM, Saunders MI, Dische S (2000) Updated data for Chen ZL, Perez S, Holmes EC, Wang HJ, Coulson WF, Wen
CHART in NSCLC: further analyses. Radiother Oncol DR, Cochran AJ (1993) Frequency and distribution of
55:86–87 occult micrometastasis in lymph nodes of patients with
Bentzen SM, Saunders MI, Dische S, Parmar MK (2001) non-small cell lung carcinoma. J Natl Cancer Inst 85:493–
Accelerated radiotherapy vs chemoradiation in non-small 498
cell lung cancer: quantifying the hazards. Radiother Oncol Chen M, Jiang G-L, Fu X-L, Wang LJ, Qian H, Chen GY, Zhao
58:91–92 S, Liu TF (2000) The impact of overall treatment time on
Black WC, Armstrong P, Daniel TM (1988) Cost effectiveness outcomes in radiation therapy for non-small cell lung
of chest CT in T1N0M0 lung cancer. Radiology 167: cancer. Lung Cancer 28:11–19
373–378 Cheung PC, Mackillop WJ, Dixon P, Brundage MD, Youssef
Blomgren H, Lax I, Naslund I, Svanstrom R (1995) Stereotactic YM, Zhou S (2000) Involved-field radiotherapy alone for
high dose fraction radiation therapy of extracranial tumors early-stage non-small-cell lung cancer. Int J Radiat Oncol
using an accelerator. Clinical experience of the first thirty- Biol Phys 48:703–710
one patients. Acta Oncol 34:861–870 Conces DJ Jr., Klink JF, Tarver RD, Moak GD (1989) T1N0M0
Bogart JA, Hodgson L, Seagren SL, Blackstock AW, Wang X, lung cancer: evaluation with CT. Radiology 170:643–646
Lenox R, Turrisi AT 3rd, Reilly J, Gajra A, Vokes EE, Cooper JFD, Pearson FG, Todd TRJ (1985) Radiotherapy alone
Green MR (2010) Phase I study of accelerated conformal for patients with operable carcinoma of the lung. Chest
radiotherapy for stage I non-small-cell lung cancer in 87:289–292
patients with pulmonary dysfunction: CALGB 39904. J Clin Cox JD, Azarnia N, Byhardt RW, Shin KH, Emami B, Pajak TF
Oncol 28:202–206 (1990) A randomized phase I/II trial of hyperfractionated
Bradley JD, Ieumwananonthachai N, Purdy JA (2002) Gross radiation therapy with total doses of 60.0 to 79.2 Gy–
tumor volume, critical prognostic factor in patients treated possible survival benefit with and gt; 69.6 Gy in favorable
with three-dimensional conformal radiation therapy for non- patients with radiation therapy oncology group stage III
small cell lung carcinoma. Int J Radiat Oncol Biol Phys nonsmall cell lung carcinoma. Report of radiation therapy
52:49–57 oncology group 83-11. J Clin Oncol 8:1543–1555
Bradley JD, Wahab S, Lockett MA, Perez CA, Purdy JA (2003) Cox JD, Pajak TF, Asbell S, Russell AH, Pederson J, Byhardt
Elective nodal failures are uncommon in medically inoper- RW, Emami B, Roach M 3rd (1993) Interruptions of high-
able patients with stage I non-small-cell lung carcinoma dose radiation therapy decrease long-term survival of
treated with limited radiotherapy fields. Int J Radiat Oncol favorable patients with unresectable non-small cell carci-
Biol Phys 56:342–347 noma of the lung: analysis of 1244 cases from 3 radiation
Bradley J, Thorstad WL, Mutic S, Miller TR, Dehdashti F, therapy oncology group (RTOG) trials. Int J Radiat Oncol
Siegel BA, Bosch W, Bertrand RJ (2004) Impact of FDG- Biol Phys 27:493–498
PET on radiation therapy volume delineation in non-small- Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the
cell lung cancer. Int J Radiat Oncol Biol Phys 59:78–86 radiation therapy oncology group (RTOG) and the European
Bradley J, Graham MV, Winter K, Purdy JA, Komaki R, Roa organization for the treatment and research of cancer
WH, Ryu JK, Bosch W, Emami B (2005) Toxicity and (EORTC). Int J Radiat Oncol Biol Phys 31:1341–1346
outcome results of RTOG 9311: a phase I-II dose-escalation Coy P, Kennelly GM (1980) The role of curative radiotherapy
study using three-dimensional conformal radiotherapy in in the treatment of lung cancer. Cancer 45:698–702
patients with inoperable non-small-cell lung carcinoma. Int De Petris L, Lax I, Sirzen F (2005) Role of gross tumor volume
J Radiat Oncol Biol Phys 61:318–328 on outcome and of dose paramerets on toxicity of patients
Braun SR, doPico GA, Olson CE, Caldwell W (1975) Low- undergoing chemoradiotherapy for locally advanced non-
dose radiation pneumonitis. Cancer 35:1322–1324 small cell lung cancer. Med Oncol 22:375–381
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 337

Detterbeck FC, Falen S, Rivera MP, Halle JS, Socinski MA Fukumoto S, Shirato H, Shimizu S, Ogura S, Onimaru R,
(2004) Seeking a home for a PET, part 2: defining the Kitamura K, Yamazaki K, Miyasaka K, Nishimura M,
appropriate place for positron emission tomography imag- Dosaka-Akita H (2002) Small-volume image-guided radio-
ing in the staging of patients with suspected lung cancer. therapy using hypofractionated coplanar and noncoplanar
Chest 125:2300–2308 multiple fields with inoperable stage I nonsmall cell lung
Dobashi K, Sugio K, Osaki T, Oka T, Yasumoto K (1997) carcinomas. Cancer 95:1546–1553
Micrometastatic P53-positive cells in the lymph nodes of Furuta M, Hayakawa K, Katano S, Saito Y, Nakayama Y,
non-small-cell lung cancer: prognostic significance. J Thorac Takahashi T, Imai R, Ebara T, Mitsuhashi N, Niibe H
Cardiovsc Surg 114:339–346 (1996) Radiation therapy for stage I-II non-small cell lung
Dosoretz DE, Katin MJ, Blitzer PH, Rubenstein JH, Salenius S, cancer in patients aged 75 years and older. Jpn J Clin Oncol
Rashid M, Dosani RA, Mestas G, Siegel AD, Chadha TT 26:95–98
(1992) Radiation therapy in the management of medically Fuwa N, Daimon T, Mitsudomi T, Yatabe Y, Kodaira T,
inoperable carcinoma of the lung: results and implications Tachibana H, Nakamura T, Kato T, Sato Y (2008)
for future treatment strategies. Int J Radiat Oncol Biol Phys Identifying patients with peropheral-type early non-small
25:3–9 cell lung cancer (T1N0M0) for whom irradiation of the
Dosoretz DE, Galmarini D, Rubenstein JH, Katin MJ, Blitzer primary focus alone could lead to successful treatment. Br J
PH, Salenius SA, Dosani RA, Rashid M, Mestas G, Hannan Radiol 81:815–820
SE (1993) Local control in medically inoperable lung Garipagaoglu M, Munley MT, Hollis D, Poulson JM, Bentel
cancer: an analysis of its importance in outcome and factors GC, Sibley G, Anscher MS, Fan M, Jaszczak RJ, Coleman
determining the probability of tumour eradication. Int J RE, Marks LB (1999) The effect of patient specific factors
Radiat Oncol Biol Phys 27:507–516 on radiation induced regional lung injury. Int J Radiat Oncol
Drzymala RE, Mohan R, Brewster L, Chu J, Goitein M, Harms Biol Phys 45:3331–3338
W, Urie M (1991) Dose–volume histograms. Int J Radiat Gauden SJ, Tripcony L (2001) The curative treatment by
Oncol Biol Phys 21:71–78 radiation therapy alone of stage I non-small cell lung cancer
Dunbar SF, Tarbell NJ, Kooy HM, Alexander E 3rd, Black PM, in a geriatric population. Lung Cancer 32:71–79
Barnes PD, Goumnerova L, Scott RM, Pomeroy SL, La Gauden S, Ramsay J, Tripcony L (1995) The curative treatment
Vally B (1994) Stereotactic radiotherapy for pediatric and by radiotherapy alone of stage I non-small cell carcinoma of
adult brain tumors: preliminary report. Int J Radiat Oncol the lung. Chest 108:1278–1282
Biol Phys 30:531–539 Ginsberg RJ, Rubinstein L (1995) A randomised study of
Edelman MJ, Gandara DR (2009) Lung Cancer. In: Casciato lobectomy versus limited resection for patients with
DA (ed) Manuel of clinical oncology, 6th edn. Lippincott T1N0 non-small cell lung cancer. Ann Thorac Surg 60:
Williams and Wilkins, Philadephia, pp 169–187 908–913
Etiz D, Marks LB, Zhou SM (2002) Infleuncew of tumor Giraud P, Antoine M, Larrouy A, Milleron B, Callard P, De
voluem on survival in patients irradiated for non-small-cell Rycke Y, Carette MF, Rosenwald JC, Cosset JM, Housset
lung cancer. Int J Radiat Oncol Biol Phys 53:835–846 M, Touboul E (2000) Evaluation of microscopic tumor
Fang LC, Komaki R, Allen P, Guerrero T, Mohan R, Cox JD extension in non-small-cell lung cancer for three-dimen-
(2006) Comparison of outcomes for patients with medically sional conformal radiotherapy planning. Int J Radiat Oncol
inoperable stage I non-small-cell lung cancer treated with Biol Phys 48:1015–1024
two-dimensional vs. three-dimensional radiotherapy. Int J Glazer GM, Orringer MB, Gross BH, Quint LE (1984) The
Radiat Oncol Biol Phys 66:108–116 mediastinum in non-small cell lung cancer. Am J Roentge-
Faria SL, Souhami L, Portelance L, Duclos M, Vuong T, Small nol 142:1101–1105
D, Freeman CR (2006) Absence of toxicity with hypofrac- Goldstraw P, Crowley J, Chansky K, Giroux DJ, Groome PA,
tionated 3-dimensional radiation therapy for inoperable, Rami-Porta R, Postmus PE, Rusch V, Sobin L, International
early stage non-small cell lung cancer. Radiat Oncol 1:42 Association for the Study of Lung Cancer International
Farrell MA, McAdams HP, Herndon JE, Patz EF Jr. (2000) Staging Committee; Participating Institutions (2007) The
Non-small cell lung cancer: FDG PET for nodal staging in IASLC lung cancer staging project: proposals for the
patients with stage I disease. Radiology 215:886–890 revision of the TNM stage groupings in the forthcoming
Firat S, Bousamra M, Gore E, Byhardt RW (2002) Comorbidity (seventh) edition of the TNM Classification of malignant
and KPS are independent prognostic factors in stage I non- tumours. J Thorac Oncol 2:706–714
small-cell lung cancer. Int J Radiat Oncol Biol Phys Graham MV, Matthews JW, Harms WB, Emami B, Glazer HS,
52:1047–1057 Purdy JA (1994) Three-dimensional radiation treatment
Flickinger JC, Kondziolka D, Lunsford LD, Coffey RJ, planning study for patients with carcinoma of the lung. Int J
Goodman ML, Shaw EG, Hudgins WR, Weiner R, Harsh Radiat Oncol Biol Phys 29:1105–1117
GR, 4th Sneed PK (1994) A multi-institutional experience Graham PH, Gebski VJ, Langlands AO (1995) Radical
with stereotactic radiosurgery for solitary brain metastasis. radiotherapy for early nonsmall cell lung cancer. Int J
Int J Radiat Oncol Biol Phys 28:797–802 Radiat Oncol Biol Phys 31:261–266
Fontanini G, Macchiarini P, Pepe S, Ruggiero A, Hardin M, Graham MV, Purdy JA, Emami B, Harms W, Bosch W,
Bigini D, Vignati S, Pingitore R, Angeletti CA (1992) The Lockett MA, Perez CA (1999) Clinical dose–volume
expression of proliferative cell nuclear antigen in paraffin histogram analysis for pneumonitis after 3D treatment for
section of peripheral node-negative non-small cell lung non-small cell lung cancer (NSCLC). Int J Radiat Oncol
cancer. Cancer 70:1520–1527 Biol Phys 45:323–329
338 B. Jeremić et al.

Grills IS, Fitch DL, Goldstein NS, Yan D, Chmielewski GW, nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys
Welsh RJ, Kestin LL (2007) Clinicopathologic analysis of 38:521–525
microscopic extension in lung adenocarcinoma: defining Jeremic B, Shibamoto Y, Acimovic LJ, Milisavljevic S (1999)
clinical target volume for radiotherapy. Int J Radiat Oncol Hyperfractionated radiotherapy for clinical stage II non-
Biol Phys 69:334–341 small cell lung cancer. Radiother Oncol 51:141–145
Haffty BG, Goldberg NB, Gerstley J, Fischer DB, Peschel RE Jeremic B, Classen J, Bamberg M (2002) Radiation therapy
(1988) Results of radical radiation therapy in clinical stage alone in technically operable, medically inoperable early
I, technically operable non-small cell lung cancer. Int J stage (I/II) non-small-cell lung cancer. Int J Radiat Oncol
Radiat Oncol Biol Phys 15:69–73 Biol Phys 5:119–130
Hara R, Itami J, Kondo T, Aruga T, Abe Y, Ito M, Fuse M, Jeremic B, Classen J, Bamberg M (2003a) Re: observation-only
Shinohara D, Nagaoka T, Kobiki T (2002) Stereotactic management of early stage, medically inoperable lung
single high dose irradiation of lung tumors under respiratory cancer. Do not do that–a loud and a clear radiotherapeutic
gating. Radiother Oncol 63:159–163 point of view. Chest 123:313–314
Hayakawa K, Mitsuhashi N, Nakajima N (1992) Radiation Jeremic B, Shibamoto Y, Milicic B, Dagovic A, Nikolic N,
therapy for stage III epidermoid carcinoma of the lung. Aleksandrovic J, Acimovic L, Milisavljevic S (2003b)
Lung Cancer 8:213–224 Impact of treatment interruptions due to toxicity on outcome
Hayakawa K, Mitsuhashi N, Saito Y, Nakayama Y, Furuta M, of patients with early stage (I/II) non-small-cell lung cancer
Sakurai H, Kawashima M, Ohno T, Nasu S, Niibe H (1999) (NSCLC) treated with hyperfractionated radiation therapy
Limited field irradiation for medically inoperable patients (Hfx RT) alone. Lung Cancer 40:317–323
with peripheral stage I non-small cell lung cancer. Lung Jeremic B, Milicic B, Acimovic L, Milisavljevic S (2005)
Cancer 26:137–142 Concurrent hyperfractionated radiotherapy and low-dose
Hayakawa K, Mitsuhashi N, Katano S, Saito Y, Nakayama Y, daily carboplatin/paclitaxel in patients with early stage (I/II)
Sakurai H, Akimoto T, Hasegawa M, Yamakawa M, Niibe non-small cell lung cancer (NSCLC). Long -term results of
H (2001) High-dose radiation therapy for elderly patients a phase II study. J Clin Oncol 23:6873–6880
with inoperable or unresectable non-small cell lung cancer. Kaskowitz L, Graham MV, Emami B, Halverson KJ, Rush C
Lung Cancer 32:81–88 (1993) Radiation therapy alone for stage I non-small cell
Hayman JA, Martel MK, Ten Haken RK, Normolle DP, Todd lung cancer. Int J Radiat Oncol Biol Phys 27:517–523
RF 3rd, Littles JF, Sullivan MA, Possert PW, Turrisi AT, Koike T, Terashima M, Takizawa T, Watanabe T, Kurita Y,
Lichter AS (2001) Dose-escalation in non-small-cell lung Yokoyama A (1998) Clinical analysis of small-sized
cancer using three-dimensional conformal radiation therapy. peripheral lung cancer. J Thorac Cardiovasc Surg 115:
Update of a phase I trial. J Clin Oncol 19:127–136 1015–1020
Heavey LR, Glazer GM, Gross BH, Francis IR, Orringer MB Komaki R, Scott C, Ettinger D, Lee JS, Fossella FV, Curran W,
(1986) The role of CT in staging radiographic T1N0M0 Evans RF, Rubin P, Byhardt RW (1997) Randomized study
lung cancer. Am J Roentgenol 146:285–290 of chemotherapy/radiation therapy combinations for favor-
Henschke CI, Wisnivesky JP, Yankelevitz DF, Miettinen OS able patients with locally advanced inoperable nonsmall cell
(2003) Small stage I cancers of the lung: genuineness and lung cancer: radiation therapy oncology group (RTOG) 92-
curability. Lung Cancer 39:327–330 04. Int J Radiat Oncol Biol Phys 38:149–155
Hof H, Herfarth K, Munter M, Hoess A, Motsch J, Wannenm- Kooy HM, Dunbar SF, Tarbell NJ, Mannarino E, Ferarro N,
acher M, Debus J (2003) Stereotactic single-dose radiother- Shusterman S, Bellerive M, Finn L, McDonough CV,
apy of stage I non-small-cell lung cancer (NSCLC). Int J Loeffler JS (1994) Adaptation of the relocatable Gill–
Radiat Oncol Biol Phys 56:335–341 Thomas–Cosman frame in stereotactic radiotherapy. Int J
Holsti LB, Vuorinen P (1967) Radiation reaction in the lung Radiat Oncol Biol Phys 30:685–691
after continuous and split-course megavoltage radiotherapy Krol AD, Aussems P, Noordijk EM, Hermans J, Leer JW
of bronchial carcinoma. Br J Radiol 40:280–284 (1996) Local irradiation alone for peripheral stage I lung
Hsie M, Morbidini-Gaffney S, Kohman LJ, Dexter E, Scalzetti cancer: could we omit the elective regional nodal irradia-
EM, Bogart JA (2007) Definitive treatment of poor-risk tion? Int J Radiat Oncol Biol Phys 34:297–302
patients with stage I lung cancer. A single institution Kupelian PA, Komaki R, Allen P (1996) Prognostic factors in
experience. J Thorac Oncol 4:69–73 the treatment of node-negative nonsmall cell lung carci-
Ishida T, Yano T, Maeda K (1991) Strategy for lymphadenop- noma with radiotherapy alone. Int J Radiat Oncol Biol Phys
athy in lung cancer 3 cm or less in diameter. Ann Thorac 36:607–613
Surg 50:708–771 Kutcher GJ, Burman C (1987) Calculation of probability
Jeremic B (2004) Incidental irradiation of nodal regions at risk factors for non-uniform normal tissue irradiation: the
during limited-field radiotherapy (RT) in dose-escalation effective volume method. Med Phys 14:487
studies in nonsmall cell lung cancer (NSCLC). Enough to Kwa SL, Theuws JC, Wagenaar A, Damen EM, Boersma LJ,
convert no-elective into elective nodal irradiation (ENI)? Baas P, Muller SH, Lebesque JV (1998) Evaluation of two
Radiother Oncol 71:123–125 dose–volume histogram reduction models for the prediction
Jeremic B (2007) Low incidence of isolated nodal failures after of radiation pneumonitis. Radiother Oncol 48:61–69
involved-field radiation therapy for non small-cell lung Lagerwaard FJ, Senan S, van Meerbeeck JP, Graveland WJ
cancer: blinded by the light? J Clin Oncol 25:5543–5554 (2002) Has 3-D conformal radiotherapy (3D-CRT) improved
Jeremic B, Shibamoto Y, Acimovic LJ, Milisavljevic S (1997) local tumour control for stage I non-small cell lung cancer?
Hyperfractionated radiotherapy alone for clinical stage I Radiother Oncol 63:151–157
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 339

Langendijk JA, Aaronson NK, ten Velde GPM, de Jong JM, and micrometastases in the lymph nodes of patients with
Muller MJ, Wouters EF (2000) Pretreatment quality of life stage I non-small cell lung cancer. J Thorac Cardiovasc
of inoperable non-small cell lung cancer patients referred Surg 114:535–543
for primary radiotherapy. Acta Oncol 39:949–958 McDonald S, Rubin P, Phillips TL, Marks LB (1995) Injury to
Lee JS, Scott C, Komaki R, Fossella FV, Dundas GS, McDonald the lung from cancer therapy: clinical syndromes, measur-
S, Byhardt RW, Curran WJ Jr. (1996) Concurrent chemora- able endpoints, and potential scoring systems. Int J Radiat
diation therapy with oral etoposide and cisplatin for locally Oncol Biol Phys 31:1187–1203
advanced inoperable non-small-cell lung cancer: radiation McGarry RC, Song G, des Rosiers P, Timmermann R (2002)
therapy oncology group protocol 91-06. J Clin Oncol Observation-only management of early stage, medically
14:1055–1064 inoperable lung cancer. Poor outcome. Chest 121:1155–
Lee S, Choi EK, Park HJ, Ahn SD, Kim JH, Kim KJ, Yoon SM, 1158
Kim YS, Yi BY (2003) Stereotactic body frame based Mizushima Y, Noto H, Sugiyama S, Kusajima Y, Yamashita R,
fractionated radiosurgery on consecutive days for primary Kashii T, Kobayashi M (1997) Survival and prognosis after
or metastatic tumors in the lung. Lung Cancer 40:309–315 pneumonectomy for lung cancer in the elderly. Ann Thorac
Lin EC, Alavi A (2009) Thoracic neoplasms. In: Lin EC, Alavi Surg 64:193–198
A (eds) PET/CT: a clinical guide, 2nd edn. Thieme, New Morita K, Fuwa N, Suzuki Y, Nishio M, Sakai K, Tamaki Y,
York, pp 142–154 Niibe H, Chujo M, Wada S, Sugawara T, Kita M (1997)
Low JSH, Koh W-Y, Yap S-P, Fong K-W (2007) Radical Radical radiotherapy for medically inoperable non-small
radiotherapy in stage I non-small cell lung cancer cell lung cancer in clinical stage I: retrospective analysis of
(NSCLC)—Singapore National Cancer Centre Expereince. 149 patients. Radiother Oncol 42:31–36
Ann Acad Med Singapore 36:778–783 Morrison R, Delley TJ, Cleland WP (1963) The treatment of
Lyman JT, Wolbarst AB (1987) Optimization of radiation carcinoma of the bronchus. A clinical trial to compare
therapy III. A method of assessing complication probabil- surgery and supervoltage radiotherapy. Lancet 1:683–684
ities from dose–volume histograms. Int J Radiat Oncol Biol Moss WT, Haddy FJ, Sweany SK (1960) Some factors altering
Phys 13:103–109 the severity of acute radiation pneumonitis: variation with
MacManus MP, Hicks RJ, Matthews JP, Hogg A, McKenzie cortisone, heparin, and antibiotica. Radiology 75:50–54
AF, Wirth A, Ware RE, Ball DL (2001) High rate of Mountain CF (1986) A new international staging system for
detection of unsuspected distant metastases by PET in lung cancer. Chest 89:225S–233S
apparent stage III non-small-cell lung cancer: implications Mountain CF (1997) Revisions in the international system for
for radical therapy. Int J Radiat Oncol Biol Phys 50:287– staging lung cancer. Chest 111:1710–1717
294 Movsas B, Scott C, Sause W, Byhardt R, Komaki R, Cox J,
Maguire PD, Sibley GS, Zhou SM, Jamieson TA, Light KL, Johnson D, Lawton C, Dar AR, Wasserman T, Roach M,
Antoine PA, Herndon JE 2nd, Anscher MS, Marks LB Lee JS, Andras JE (1999) The benefit of treatment
(1999) Clinical and dosimetric predictors of radiation- intensification is age and histology-dependent in patients
induced esophageal toxicity. Int J Radiat Oncol Biol Phys with locally advanced non-small cell lung cancer (NSCLC):
45:97–103 a quality-adjusted survival analysis of radiation therapy
Maguire PD, Marks LB, Sibley GS, Herndon JE 2nd, Clough oncology group (RTOG) chemoradiation studies. Int J
RW, Light KL, Hernando ML, Antoine PA, Anscher MS Radiat Oncol Biol Phys 45:1143–1149
(2001) 73.6 Gy and beyond: hyperfractionated, accelerated Movsas B, Scott C, Sause W (2000) Age dramatically impacts
radiotherapy for non-small-cell lung cancer. J Clin Oncol on the quality-adjusted time without symptoms or toxicity
19:705–711 (Q_Twist) in locally advanced non-small cell lung cancer
Mah K, Van Dyk J, Keane T, Poon PY (1987) Acute radiation- (LA-NSCLC)—a radiation therapy oncology group
induced pulmonary damage: a clinical study on the response (RTOG) analysis. Proc 42nd Ann Meet Am Soc Ther
to fractionated radiation therapy. Int J Radiat Oncol Biol Radiol Oncol (Abstract 92)
Phys 13:179–188 Nagata Y, Negoro Y, Aoki T, Mizowaki T, Takayama K,
Marom EM, McAdams HP, Erasmus JJ, Goodman PC, Culhane Kokubo M, Araki N, Mitsumori M, Sasai K, Shibamoto Y,
DK, Coleman RE, Herndon JE, Patz EF Jr. (1999) Staging Koga S, Yano S, Hiraoka M (2002) Clinical outcomes of 3D
non-small cell lung cancer with whole-body PET. Radiol- conformal hypofractionated single high-dose radiotherapy
ogy 212:803–809 for one or two lung tumors using a stereotactic body frame.
Martel MK, Ten Haken RK, Hazuka MB, Turrisi AT, Fraass Int J Radiat Oncol Biol Phys 52:1041–1046
BA, Lichter AS (1994) Dose–volume histogram and 3-D Naruke T, Goya T, Tsuchiya R, Suemasu K (1988) Prognosis
treatment planning evaluation of patients with pneumonitis. and survival in resected lung carcinoma based on the new
Int J Radiat Oncol Biol Phys 28:575–581 international staging system. J Thorac Cardiovasc Surg
Martel MK, Sahijdak WM, Hayman JA (1999) Incidental dose 96:440–447
to clinically negative nodes from conformal treatment fields Newlin HE, Iyengar M, Morris CG, Olivier K (2009)
for nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys Unresectable squamous cell carcinoma of the lung: an
45(3Suppl):244 Abstract outcomes study. Int J Radiat Oncol Biol Phys 74:370–376
Martini N, Beattie E (1977) Results of surgical treatment in Noordijk EM, Poest Clement Evd, Hermans J, Wever AM, Leer
stage I lung cancer. J Thorac Cardiovasc Surg 74:499–505 JW (1988) Radiotherapy as an alternative to surgery in
Maruyama R, Sugio K, Mitsudomi T, Saitoh G, Ishida T, elderly patients with respectable lung cancer. Radiother
Sugimachi K (1997) Relationship between early recurrence Oncol 13:83–89
340 B. Jeremić et al.

Oetzel D, Schraube P, Hensley F, Sroka-Perez G, Menke M, following radiation therapy alone for stage I non-small cell
Flentje M (1995) Estimation of pneumonitis risk in three- lung cancer. Int J Radiat Oncol Biol Phys 19:9–13
dimensional treatment planning using dose–volume histo- Saunders M, Dische S, Barrett A, Harvey A, Griffiths G, Palmar
gram analysis. Int J Radiat Oncol Biol Phys 33:455–460 M (1999a) Continuous hyperfractionated accelerated radio-
Onimaru R, Shirato H, Shimizu S, Kitamura K, Xu B, therapy (CHART) versus conventional radiotherapy in non-
Fukumoto S, Chang T-C, Fujita K, Oita M, Miyasaka K, small cell lung cancer: mature data from the randomised
Nishimura M, Dosaka-Akita H (2003) Tolerance of organs multicentre trial. Radiother Oncol 52:137–148
at riskin small-volume, hypofractionated, image-guided Saunders CA, Dussek JE, O0 Doherty MJ, Maisey MN (1999b)
radiotherapy for primary and metastatic lung cancers. Int J Evaluation of fluorine-18-fluorodeoxyglucose whole body
Radiat Oncol Biol Phys 56:126–135 positron emission tomography imaging in the staging of
Ono R, Egawa S, Suemasu K, Sakura M, Kitagawa T (1991) lung cancer. Ann Thorac Surg 76:790–797
Radiotherapy in inoperable stage I lung cancer. Jpn J Clin Sause WT, Scott C, Taylor S, Johnson D, Livingston R,
Oncol 21:125–128 Komaki R, Emami B, Curran WJ, Byhardt RW, Turrisi AT
Passlick B, Izbicki JR, Kubuschok B, Thetter O, Pantel K (1995) Radiation therapy oncology group 88–08 and eastern
(1996) Detection of disseminated lung cancer cells in lymph cooperative oncology group 4588: preliminary results of a
nodes: impact on staging and prognosis. Ann Thorac Surg phase III trial in regionally advanced, unresectable nonsmall
61:177–183 cell lung cancer. J Natl Cancer Inst 87:198–205
Pastorino U, Andreola S, Tagliabue E, Pezzella F, Incarbone M, Sawyer TE, Bonner JA, Gould PM, Deschamps C, Lange CM,
Sozzi G, Buyse M, Menard S, Pierotti M, Rilke F (1997) Li H (1999) Predictors of subclinical nodal involvement in
Immunocytochemical markers in stage I lung cancer: clinical stages I and II non-small cell lung cancer. Impli-
relevance to prognosis. J Clin Oncol 15:2858–2865 cations in the inoperable and three-dimensional dose-
Pemberton LS, Din OS, Fisher PM, Hatton MQ (2009) escalation settings. Int J Radiat Oncol Biol Phys 43:965–
Accelerated radical radiotherapy for non-small cell lung 970
cancer using two common regimens: a single-centre retro- Senan S, Burgers S, Samson MJ, van Klaveren RJ, Oei SS, van
spective study on outcome. Clin Oncol 21:161–167 Sornsen de Koste J, Voet PW, Lagerwaard FJ, Maarten van
Potosky AL, Saxman A, Wallace RB (2004) Population Haarst J, Aerts JG, van Meerbeeck JP (2002) Can elective
variations in the initial treatment of non-small-cell lung nodal irradiation be omitted in stage III non-small-cell lung
cancer. J Clin Oncol 22:3261–3268 cancer ? Analysis of recurrences in a phase II study of
Prasad SC (1978) Relation between tolerance dose and induction chemotherapy and involved-field radiotherapy. Int
treatment field size in radiation therapy. Med Phys 5:430– J Radiat Oncol Biol Phys 54:999–1006
433 Sibley GS, Jamieson TA, Marks LB, Anscher MS, Prosnitz LR
Raz DJ, Zell JA, Ou SH, Gandara DR, Anton-Culver H, Jablons (1998) Radiotherapy alone for medically inoperable stage I
DM (2007a) Natural history of stage I non-small cell lung non-small-cell lung cancer: the Duke experience. Int J
cancer: implications for early detection. Chest 132:193–199 Radiat Oncol Biol Phys 40:149–154
Raz DJ, Zell JA, Ou SH (2007b) Natural history of stage I non- Slebos RJ, Kibbelaar RE, Dalesio O, Kooistra A, Stam J,
small-cell lung cancer: implications for early detection. Meijer CJ, Wagenaar SS, Vanderschueren RG, van Zan-
Chest 132:193–199 dwijk N, Mooi WJ (1990) K-ras oncogene activation as
Robertson JM, Ten Haken RK, Hazuka MB, Turrisi AT, Martel prognostic marker in adenocarcinoma of lung. N Engl J
MK, Pu AT, Littles JF, Martinez FJ, Francis IR, Quint LE, Med 323:561–566
Lichter AS (1997) Dose escalation for non-small cell lung Slotman BJ, Karim ABMF (1994) Curative radiotherapy for
cancer using conformal radiation therapy. Int J Radiat Oncol technically operable stage I nonsmall cell lung cancer. Int J
Biol Phys 37:1079–1085 Radiat Oncol Biol Phys 29:33–37
Rosenthal SA, Curran WJ Jr., Herbert SH, Hughes EN, Sandler Slotman BJ, Antonisse IE, Njo KH (1996) Limited field
HM, Stafford PM, McKenna WG (1992) Clinical stage II irradiation in early stage (T1–2 N0) non-small cell lung
non-small cell lung cancer treated with radiation therapy cancer. Radiother Oncol 41:41–44
alone. The significance of clinically staged ipsilateral hilar Smart J (1966) Can lung cancer be cured by irradiation alone?
adenopathy (N1 disease). Cancer 70:3410–3417 JAMA 195:1034–1035
Rosenzweig KE, Sim SE, Mychalczak B, Braban LE, Schindel- Soliman H, Cheung P, Yeung L, Poon I, Balogh J, Barbera L,
heim R, Leibel SA (2001) Elective nodal irradiation in the Spayne J, Danjoux C, Dahele M, Ung Y (2011) Accelerated
treatment of non-small-cell lung cancer with three-dimen- hypofractionated radiotherapy for early-stage non-small-cell
sional conformal radiation therapy. Int J Radiat Oncol Biol lung cancer: long-term results. Int J Radiat Oncol Biol Phys
Phys 50:681–685 79:459–465
Rubin P, Casarett GW (1968) Clinical radiation pathology. Sturm V, Kober H, Hover K-H, Schlegel W, Boesecke R,
Philadelphia, Saunders, pp 423–470 Pastyr O, Hartmann GH, Schabbert S, zum Winkel K,
Sandhu AP, Messer K, Fuster MM, Ahmad E, Pu M, Kunze S (1987) Stereotactic percutaneous single dose
Bazhrenova L, Rose M, Seagren S (2009) Definitive irradiation of brain metastasis with a linear accelerator. Int
radiation therapy for stage I non-small-cell lung carcinoma: J Radiat Oncol Biol Phys 13:279–282
institutional experience with contemporary conformal plan- Suzuki K, Nagai K, Yoshida J, Nishimura M, Takahashi K,
ning. Clin Lung Cancer 10:433–437 Nishiwaki Y (1999) Clinical predictors of N2 disease in the
Sandler HM, Curran WJ Jr., Turrisi AT III (1990) The influence setting of a negative computed tomographic scan in patients
of tumor size and pre-treatment staging on outcome with lung cancer. J Thorac Cardiovasc Surg 117:593–598
Conventional Radiation Therapy in Early Stage Non-small-cell Lung Cancer 341

Suzuki K, Nagai K, Yoshida J, Nishimura M, Nishiwaki Y medically inoperable non-small-cell lung cancer. J Med
(2001) Predictors of lymph node and intrapulmonary Imag Radiat Oncol 54:554–561
metastasis in clinical stage IA non-small cell lung carci- Whittle J, Steinberg EP, Anderson G, Herbert R (1991) Use of
noma. Ann Thorac Surg 72:352–356 Medicare claims data to evaluate outcomes in elderly
Takizawa T, Terashima M, Koike T, Watanabe T, Kurita Y, patients undergoing lung resection for lung cancer. Chest
Yokoyama A, Honma K (1998) Lymph node metastasis in 100:729–734
small peripheral adenocarcinoma of the lung. J Thorac Whyte RI, Crownover R, Murphy MJ, Martin DP, Rice TW,
Cardiovasc Surg 116:276–280 DeCamp M Jr., Rodebaugh R, Weinhous MS, Le Q-T
Talton BM, Constable WC, Kersh CR (1990) Curative (2003) Stereotactic radiosurgery for lung tumors: prelimin-
radiotherapy for technically operable stage I nonsmall cell ary results of a phase I trial. Ann Thorac Surg 75:1097–
lung cancer. Int J Radiat Oncol Biol Phys 19:15–21 1101
Tateishi M, Ishida T, Mitsudomi Kaneko S, Sugimachi K Williams TE, Thomas CR Jr., Turrisi AT III (2000) Better
(1991) Prognostic value of c-erB-2 protein expression in radiation treatment of non-small cell lung cancer using new
human lung adenocarcinoma and squamous cell carcinoma. techniques without elective nodal irradiation. Semin Rad
Eur J Cancer 27:1372–1375 Onc 10:315–323
Timmerman R, Papiez L, McGarry R, Likes L, DesRosiers C, Willner J, Baier K, Caragiani E (2002) Dose, volume and tumor
Frost S, Williams M (2003) Extracranial stereotactic control prediction in primary radiotherapy of non-small-cell
radioablation. Results of a phase I study in medically lung cancer. Int J Radiat Oncol Biol Phys 52:382–389
inoperable stage I non-small cell lung cancer. Chest 123: Wu J, Ohta Y, Minato H, Tsunezuka Y, Oda M, Watanabe Y,
1946–1955 Watanabe G (2001) Nodal occult metastasis in patients with
Timmerman R, Paulus R, Galvin J, Michalski J, Straube W, peripheral lung adenocarcinoma of 2.0 cm or less I
Bradley J, Fakiris A, Bezjak A, Videtic G, Johnstone D, ndiameter. Ann Thorac Surg 71:1772–1777
Fowler J, Gore E, Choy H (2010) Stereotactic body Yom SS, Liao Z, Liu HH, Tucker SL, Hu CS, Wei X, Wang X,
radiation therapy for inoperable early stage lung cancer. Wang S, Mohan R, Cox JD, Komaki R (2007) Initial
JAMA 303:1070–1076 evaluation of treatment-related pneumonitis in advanced-
Uematsu M, Shioda A, Tahara K, Fukui T, Yamamoto F, stage non-small-cell lung cancer patients treated with
Tsumatori G, Ozeki Y, Aoki T, Watanabe M, Kusano S concurrent chemotherapy and intensity-modulated radio-
(1998) Focal, high-dose, and fractionated modified stereo- therapy. Int J Radiat Oncol Biol Phys 68:94–102
tactic radiation therapy for lung carcinoma patients. A Yu HM, Liu YF, Yu JM, Liu J, Zhao Y, Hou M (2008)
preliminary experience. Cancer 82:1062–1070 Involved-field radiotherapy is effective for patients 70 years
Uematsu M, Shioda A, Suda A, Fukui T, Ozeki Y, Hama Y, old or more with early stage non-small cell lung cancer.
Wong JR, Kusano S (2001) Computed tomography-guided Radiother Oncol 87:29–34
frameless stereotactic radiotherapy for stage I non-small- Yuan S, Sun X, Li M (2007) A randomized study of involved
cell lung cancer: a 5 year experience. Int J Radiat Oncol field irradiation versus elective nodal irradiation in com-
Biol Phys 51:666–670 bination with concurrent chemotherapy for inoperable
Underberg RW, Lagerwaard FJ, Slotman BJ, Cuijpers JP, stage III nonsmall cell lung cancer. Am J Clin Oncol 30:
Senan S (2005) Use of maximum intensity projections 239–244
(MIP) for target volume generation in 4DCT scans for lung Zhang HX, Yin WB, Zhang LJ, Yang ZY, Zhang ZX, Wang M,
cancer. Int J Radiat Oncol Biol Phys 63:253–260 Chen DF, Gu XZ (1989) Curative radiotherapy of early
Varlotto JM, Shieeve DC, Alexander E III, Kooy HM, Black operable non-small cell lung cancer. Radiother Oncol
PM, Loeffler JS (1996) Fractionated stereotactic radiother- 14:89–94
apy for the treatment of acoustic neuromas: preliminary Zhao L, West BT, Hayman JA, Lyons S, Cease K, Kong F-M
results. Int J Radiat Oncol Biol Phys 36:141–145 (2007) High radiation dose may reduce the negativ effect of
Watkins JM, Wahlquist AE, Zauls AJ, Fields EC, Garrett- large gross tumor volume in patients with medically
Mayer E, Aguero EG, Silvestri GA, Sharma AK (2010) inoperable early-stage non-small cell lung cancer. Int J
High-dose fractionated radiotherapy to 80 Gy for stage I-II Radiat Oncol Biol Phys 68:103–110
Stereotactic Ablative Radiotherapy
for Early Stage Lung Cancer
John H. Heinzerling and Robert D. Timmerman

Contents Abstract
Stereotactic ablative radiotherapy (SABR), also
1 Introduction.............................................................. 343 known as stereotactic body radiation therapy
2 Techniques and Technological Advances (SBRT) utilizes advanced techniques of immobi-
in SABR for Lung Tumors..................................... 344 lization, image guidance, and unique field arrange-
2.1 Patient Positioning and Immobilization.................... 345 ments to deliver precise, oligofractionated
2.2 Daily Imaging for Patient Repositioning Prior
radiotherapy to a variety of tumor types. SABR
to Treatment............................................................... 346
2.3 Tumor Motion Assessment ....................................... 346 has been established as a technologically innova-
2.4 Tumor Motion Control .............................................. 347 tive therapy for early stage non-small cell lung
2.5 Target Volume Delineation and Treatment cancer (NSCLC) and has emerged as the standard
Planning ..................................................................... 347
treatment option for medically inoperable patients
3 Radiobiological Aspects of SABR.......................... 350 through utilization of prospective, multi-institu-
3.1 Tumor Biology .......................................................... 350 tional trials. Recent trials continue to evaluate the
3.2 Normal Tissue Radiobiology and Tolerance............ 351
role of SABR in the medically operable and
4 Clincal Results in Primary Lung Cancer............. 353 borderline operable population, and will compare
4.1 Medically Inoperable Patients with Early
surgical resection and SABR as treatment modal-
Stage Lung Cancer .................................................... 353
4.2 Medically Operable Patients ..................................... 356 ities in these patients. This chapter reviews the
techniques utilized in SABR, the evidence for use
5 Toxicity...................................................................... 357
of SABR in early stage lung cancer, its extension
6 Conclusions ............................................................... 358 of use to medically operable patients, and the
References.......................................................................... 358 toxicities associated with this emerging technique.

1 Introduction
R. D. Timmerman (&)
Stereotactic radiosurgery first emerged as a technique
Effie Marie Cain Distinguished Chair
in Cancer Therapy Research, in the 1950s created by Lars Leskell to treat intra-
Department of Radiation Oncology, cranial neoplasms using single large dose per fraction
University of Texas Southwestern Medical Center, treatments (Leskell 1951). These treatments were a
5801 Forest Park Road, Dallas, TX 75390-9183, USA
fundamental change from the perception of conven-
e-mail: robert.timmerman@utsouthwestern.edu
tionally fractionated radiotherapy (CFRT) classically
J. H. Heinzerling
used to exploit the radiobiological differences between
Department of Radiation Oncology,
University of Texas Southwestern Medical Center, normal and neoplastic tissues. Leskell established the
Dallas, TX, USA principles of immobilization and precise targeting

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 343
DOI: 10.1007/174_2011_258, Ó Springer-Verlag Berlin Heidelberg 2011
344 J. H. Heinzerling and R. D. Timmerman

utilizing a frame with fiducial markers to define an fraction treatments possible despite their profound
intracranial coordinate system that allowed treatment potency. These methods have been extended to treat
guidance, patient immobilization, and facilitated various primary and metastatic tumors including lung,
accurate dose delivery with maximization of normal liver, pancreas, kidney, adrenal gland, and prostate
tissue sparing. Based on the success of intracranial (Rusthoven et al. 2009a, b; Timmerman et al. 2010a,
radiosurgery, pioneers such as Henric Blomgren, b; Madsen et al. 2007; Koong et al. 2004; Chawla
Ingmar Lax, and Minoru Uematsu developed tech- et al. 2009; Timmerman et al. 2007a, b).
nologies that would allow similar treatment accuracy An estimated 222,000 new cases of lung cancer
for targets within the body (Blomgren et al. 1995; Lax were diagnosed in 2010, with about 30% of patients
et al. 1994; Uematsu et al. 1998). While tumors within presenting with early stage disease. The majority of
the skull have no additional movement once the skull these patients undergo surgical resection with 5 year
has been immobilized, body tumors are subject to survival rates of approximately 60–70% (Naruke et al.
forces within the body such as respiratory breathing, 1988; Nesbitt et al. 1995). While surgical resection is
cardiac contraction, and gastrointestinal peristalsis. the standard therapy in these patients, many patients
Thus, researchers from Sweden extended patient cannot tolerate surgery because of comorbidities
immobilization and fiducial targeting to the body related to lung and cardiac function. In the past, these
through construction of a body frame, and attempted patients were treated with conventional radiation typi-
to reduce internal motion of targets related to respi- cally given to a dose of approximately 45–66 Gy in
ration in order to allow high doses of radiation to be fractions of 1.8–2 Gy over 6 weeks, resulting in a
administered extracranially. New dosimetric methods 5 year survival of approximately 10–30% (Kaskowitz
were created to use multiple, non-coplanar beams with et al. 1993; Wisnivesky et al. 2005). Exploration of high
compact aperture dimensions to mimic the conver- dose per fraction treatments for primary lung cancer
gence of beams seen in Gamma KnifeÒ treatments. has now shown high rates of local control and survival
These methods to treat body tumors have collectively more comparable to surgical series. The tech-niques
been termed stereotactic ablative radiotherapy utilized in SABR treatments, the unique biologic
(SABR) (Loo et al. 2011), also known as stereotactic effects, the associated toxicity, and its careful explo-
body radiation therapy (SBRT) (Timmerman et al. ration through prospective clinical trials in patients
2003a, b). SABR is defined by the American College with early stage lung cancer will be discussed below.
of Radiology (ACR) and American Society of Ther-
apeutic Radiology and Oncology (ASTRO) as the use
of very large dose per fraction, and specific guidelines 2 Techniques and Technological
have been established for treatment implementation Advances in SABR for Lung Tumors
and quality assurance (Potters et al. 2004).
SABR is radiobiologically unique from CFRT. Safe and effective SABR treatments require ensuring
SABR treatments cause dramatic effects within tar- accuracy and precision achieved by reliable and
geted tissues both disruption of cell division and reproducible patient immobilization; daily image
function, leading to the associated term ‘‘ablative’’ guidance for precise repositioning prior to treatment;
radiotherapy. An order of magnitude of higher frac- assessment and accounting for tumor and organ
tional doses of 10–20 Gy compared to conventional motion consistently between planning and treatment;
fraction doses of 1.8–2 Gy lead to biologic ablation of and the use of multiple, non-coplanar treatment fields
cells leaving them dysfunctional not only to divide, to ensure adequate target coverage with rapid fall off
but also to perform other cellular functions. Normal of dose to surrounding normal tissues. These princi-
tissue toxicity is also affected by these ablative ples allow the reduction of normal tissue exposure to
effects, and thus it is essential to avoid unnecessary high and intermediate doses, minimizing the proba-
treatment of normal tissue surrounding the target. bility of normal tissue complications while ensuring
Improvements in imaging, targeting, on board image accurate high dose delivery to the target. Because of
guidance, dosimetric methods, and radiation delivery the proximity several critical structures including
devices have allowed increased sparing of surround- normal lung tissue, bronchi, chest wall, esophagus,
ing normal tissues and made these high dose per heart, brachial plexus, and spinal cord, realization of
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 345

Fig. 2 Example of frame system, the elekta stereotactic body


Fig. 1 Example of fiducial system in a stereotactic body frame frame showing external coordinate system, vacuum pillow, and
with coordinates corresponding to target location integrated respiratory motion control with abdominal
compression

these SABR principles is of greatest importance to that of brain radiosurgery headframes that are rigidly
ensure safe treatment with minimal toxicity while attached and registered to the target. Fiducials
achieving adequate tumor control. For CFRT, are simply reliable ‘‘markers’’ whose position can be
emphasis on correlation of the static treatment plan to consistently and confidently correlated to both the
actual treatment is less critical than for SABR because target (tumor) and treatment device (frame) (Fig. 1).
of the large margins placed around the target, the ‘‘Frame’’ systems provide both immobilization and
homogeneous dose distributions, and the more a fiducial system that can approximate initial target
‘‘forgiving’’ fractionation typically seen with limited localization independently from other image guidance
field CFRT. This, however, is not the case for SABR systems (e.g., room lasers and cone-beam CT), which
treatments, and all effort should be made to ensure the is then enhanced and adjusted by utilizing image
accuracy of equipment used in simulation, planning, guidance as described below (Lax et al. 1994; Yenice
and treatment delivery. Recently, the American et al. 2003; Herfarth et al. 2001; Wang et al. 2006). In
Association of Physicists in Medicine (AAPM) pub- addition, these systems often integrate motion control
lished their best practice guidelines for SABR treat- techniques such as abdominal compression (Fig. 2).
ments including equipment and QA procedures ‘‘Frameless’’ systems rely on the combination of
(Benedict et al. 2010). Many of the technological markers and imaging methods to effectively relocate a
advances within radiation therapy have been devel- reference position within the patient. For example,
oped to allow increased safety of oligofractionated implanted fiducial seeds within the tumor can be
treatment and will be discussed here. accurately relocated on a daily basis using imaging
techniques (Murphy 1997; Chang et al. 2004; Wulf
et al. 2000; Fuss et al. 2004). Newer technologies
2.1 Patient Positioning used in SABR treatments include electromagnetic
and Immobilization transponders that can be tracked throughout treatment
to ensure accurate positioning during the entire
Because SABR treatments are often longer than treatment course (Balter et al. 2005).
conventional radiotherapy treatment sessions, con- As with any system, assessment must be made by
sistent, reproducible, and comfortable patient immo- each institution on its ability to achieve accuracy in its
bilization is essential for ensuring treatment accuracy. primary objective: consistent patient immobilization
Several available systems designed for stereotactic and target localization. Staff training and quality
patient positioning and immobilization are currently assurance programs are essential for proper implica-
commercially available and include body frames, tion of any device, no matter its claimed accuracy.
vacuum cushions, and thermal plastic restraints. Some Since no one system has been shown to be superior,
of these devices feature a fiducial system mimicking achieving patient comfort that avoids positions in
346 J. H. Heinzerling and R. D. Timmerman

Fig. 3 Example of CBCT to validate target position prior to SBRT lung treatment. After realignment of CBCT to the planning
CT, couch shifts are shown in the bottom right and are applied prior to treatment

which the patient fights gravity or requires cumber- prior to treatment (Wang et al. 2007), but improve-
some positions for the patient to support is of utmost ments are being made including respiratory-correlated
importance when incorporating SABR into any radi- CBCT or 4D CBCT to attempt to characterize motion
ation oncology program. on a daily basis immediately prior to treatment (Sonke
et al. 2005).
Advanced image guidance, typically including
2.2 Daily Imaging for Patient mounted in-room kV imagers, has allowed monitor-
Repositioning Prior to Treatment ing of target or fiducial position during treatment as
well (Verellen et al. 2003; Chang et al. 2003; Shirato
Image guidance provides target localization and vali- et al. 2000). This has minimized uncertainty associ-
dation of patient position prior to treatment. Typically ated with external reference points and has allowed
this is performed with computed tomography (CT) accurate internal tumor localization in near real-time.
(CT on rails), or cone-beam CT (CBCT) incorporated
into the treatment unit (Jaffray et al. 2002) (Fig. 3).
Because lung tumors are very distinct on CT scan, pre- 2.3 Tumor Motion Assessment
treatment reference volumes can be registered to the
image and adjustments can be reliably made on the Because of their proximity to moving organs such as
day of treatment. Appropriate adjustments are made the diaphragm and heart, lung tumors have significant
typically with couch shifts to align pretreatment target motion up to 5 cm (Chen et al. 2001; Mageras et al.
position to the newly acquired CT data. CBCT scans 2004). This motion can vary quite significantly
are often slow in acquisition time and thus are more depending on tumor location as well as patient to
like slow CT to estimate respiratory associated motion patient (Heinzerling et al. 2008; Liu et al. 2007;
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 347

Fig. 4 Example of target definition on fast, spiral CT (a) compared to maximum intensity projection (MIP) from 4DCT
(b) showing underestimation of target excursion on fast CT

Stevens et al. 2001). Classic 3D imaging with fast dampen tumor motion include abdominal compres-
spiral CT studies can misrepresent target position by sion, which places a pressure device on the abdomen
obtaining images of the tumor in a specific phase of to dampen the motion of the diaphragm (Lax et al.
the respiratory cycle, leading to extreme errors in 1994; Wulf et al. 2000; Heinzerling et al. 2008;
target position definition (Chen et al. 2004) (Fig. 4). Negoro et al. 2001). Other techniques such as deep
Thus, a personalized assessment for each patient is inspiration breath-hold or active breathing control
required to characterize tumor motion. This was (ABC) arrest or freeze the tumor in a reproducible
typically achieved in the past with fluoroscopy and position within the respiratory cycle (Yin et al. 2001;
more recently with 4-D computed tomography O’Dell et al. 2002; Murphy et al. 2002). Respiratory
(4DCT) (Fig. 5), during which several CT images are gating utilizes respiratory cycle monitoring combined
obtained over multiple respiratory cycles and corre- with a surrogate to trigger delivery of radiation during
lated with a surrogate for breathing motion. Images a specific segment of the respiratory cycle (expiration
are then reconstructed into several respiratory phases or inspiration) (Vedam et al. 2001; Kimura et al.
(typically 10) to show anatomy through an entire 2004; Wang et al. 2001; Kini et al. 2003) (Fig. 6).
respiratory cycle. Finally, tumor tracking systems actually move the
Other techniques to quantify target motion include radiation beam path to follow the motion of the tumor
slow CT (Lagerwaard et al. 2001), breath-hold tech- (Kuriyama et al. 2003; Sharp et al. 2004; Schweikard
niques (Mageras and Yorke 2004), and respiratory- et al. 2004; Shirato et al. 1999). Regardless of the
correlated PET-CT or MRI (Kooch et al. 2004). If method used, careful assurance of accuracy, repro-
tumor motion exceeds 5–10 mm, tumor motion con- ducibility, as well as prudent implementation of
trol as described below is recommended so as to avoid motion data into treatment planning is essential for
excess toxicity. precise treatment delivery.

2.4 Tumor Motion Control 2.5 Target Volume Delineation


and Treatment Planning
Several methods exist to control tumor motion and
include techniques to reduce tumor motion (damp- Target volumes are defined using mechanisms that
ening), correlate treatment with tumor position (gat- quantify both tumor volume as well as tumor motion
ing), or track the tumor position (chasing). Ways to such as 4-dimensional CT described above. The gross
348 J. H. Heinzerling and R. D. Timmerman

Fig. 5 4DCT in the coronal plane showing a T1 tumor in b the poor correlation of tumor and diaphragm motion between
inhalation, exhalation, and maximum intensity projection (MIP) inhalation and exhalation (i.e., the diaphragm moves consider-
with lines demarcating a the tumor cranio-caudal motion ably more than the tumor)
envelop (also known as the internal target volume, ITV), and

doses to a minimum. An internal target volume or


ITV can also be delineated based on the volume
needed to encompass tumor motion. An additional
margin is then added to the defined volume to account
for daily setup error and machine tolerances, which
constitutes the planning treatment volume (PTV).
SABR requires conformal dose distributions that
allow rapid dose fall-off outside of the tumor volume.
This is accomplished by utilizing beams that are
highly shaped to the target allowing sharp collimation
of beam fluence outside of the target and using mul-
Fig. 6 Example of a respiratory gating tracing in which the breath- tiple beams (typically 10–15) or large angle arc
ing signal (top) is correlated with beam on/off status (bottom)
rotations (Cardinale et al. 1999; Liu et al. 2006;
Papiez et al. 2003). Use of non-opposing beams is
tumor volume (GTV) is typically defined on CT in encouraged to ovoid overlap of dose at points of
lung windowing. In certain cases, when the tumor is entrance and exit. In most circumstances, non-copla-
hard to differentiate from surrounding normal struc- nar beams are also preferred to provide more isotropic
tures, MRI or PET/CT can be useful to accurately dose fall-off in all directions. Typical beam arrange-
determine tumor volume. A good example of this is in ments for lung SBRT is seen in Fig. 8.
lung tumors adjacent to the chest wall or associated The resulting dose distribution seen in Fig. 9 shows
with lung atelectasis or consolidation (Fig. 7). In relatively isotropic dose fall-off in all directions.
these situations, PET/CT can be helpful for delineat- Reducing high dose spillage outside of the
ing targets more precisely. intended treatment volume is critical for preventing
Typically in SABR, like in brain radiosurgery, the normal tissue toxicity of organs at risk. Ablation is
GTV is equal to the clinical target volume (CTV), likely to occur not only in the target itself, but also
keeping the volume of normal tissues exposed to high in the shell of normal tissue immediately outside of
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 349

Fig. 7 PET/CT showing primary lung tumor with high SUV (a, b) with distal consolidation showing low SUV uptake (c, d). Use of
PET/CT for tumor delineation in this case allows decreased target volume and better sparing of normal tissue dose

Fig. 8 Typical non-coplanar beam arrangement for SABR treatment of right (a) and left (b) sided lung tumors
350 J. H. Heinzerling and R. D. Timmerman

20 Gy (V20) is quite low (\5%) compared to what is


normally seen in CFRT (20–35%). Yet, because this
dose is given in 3 fractions, using classic parameters
such as V20 can be misleading and lead to excessive
toxicity.

3 Radiobiological Aspects of SABR

3.1 Tumor Biology

Fig. 9 Dose distribution of left sided primary lung tumor Conventional understanding of tumor radiobiology
showing isotropic dose fall-off with non-coplanar beam has been mostly obtained through experimentation of
arrangement
lower doses per fraction when compared to SABR,
which involves very high doses in few fractions.
the PTV. Thus, the high dose spillage outside of the Because of these origins, traditional radiobiology has
target is directly related to the toxicity that can occur been challenged when applied to ablative, oligo-
with SABR. Tubular structures within the lung can fractionated treatments. Typically, the cell survival
be obliterated and cause subsequent downstream curve for ionizing radiation has been used to
effects such as atelectasis or consolidation. Because describe the surviving fraction versus dose, and
of this, SABR protocols commonly define criteria classically has been illustrated by the linear-qua-
related to the conformality of target dose and the dratic (LQ) formula (Hall et al. 1972; Rossi and
compactness of intermediate dose. Examples of this Kellerer 1972) (Fig. 11).
include evaluations of parameters such as ‘‘R50’’ With CFRT, daily doses ranging from 1.5 to
defined as the ratio of the 50% prescription isodose 3.0 Gy are used which occur on the ‘‘shoulder’’ of the
volume to the PTV, and ‘‘D2 cm’’defined as the survival curve, allowing cells to be able to repair
maximum dose 2 cm from the PTV in any direction. some of the radiation damage. Higher fractional doses
These parameters, which change with PTV dimen- ([3 Gy) occur on the linear portion of the curve
sions, allow evaluation of intermediate dose spillage predicting a greater proportion of cell kill. Several
that can cause more global organ damage. Confor- authors have challenged the LQ model for daily doses
mality index or the ratio of the prescription isodose beyond the shoulder (6–8 Gy), claiming that it grossly
volume to the PTV volume should be kept below 1.2 overestimates cell kill in this range (Park et al. 2008;
and typical PTV coverage should be 95–100% with Marks 1995; Guerrero and Li 2004). Several modifi-
99% of the PTV covered by 90% of prescription cations have been proposed to correctly model SABR
dose. In addition, normal tissue dose constraints are dose response curves, including utilization of the
being developed and modified based on existing data older multitarget model to ascertain a ‘‘single fraction
from patients treated with SABR to help predict equivalent dose’’ for oligofractionated treatments
toxicity based on dose-volume relationships. These ranging from 7–10 Gy, thus eliminating any influence
constraints specifically relate to total dose, fraction- of the classic LQ model (Park et al. 2008; Marks
ation, and volume of specific normal tissues. 1995). Another proposal modifies the existing LQ
Because dose-volume relationships in the setting of model by incorporating aspects of the lethal-poten-
hypofractionation are not well understood, these tially lethal model, which accounts for ongoing radi-
constraints are frequently modified based on patient ation repair processes that occur during radiation
outcome data in ongoing multicenter trials evaluat- exposure (Guerrero and Li 2004). Regardless of the
ing SABR. Figure 10 shows an example DVH from proposal, the predicted tumor cell kill becomes
the same case shown in Fig. 9. significantly different when compared to the LQ
Notice that despite the high prescription dose model, preventing overestimation of cell kill for large
(60 Gy in 3 fractions), the volume of lung getting fraction sizes used in SABR.
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 351

Fig. 10 Dose volume


histogram from SABR
primary lung treatment to
60 Gy in 3 fractions showing
low V20 (\5%)

These newly proposed models have created effects of SABR such as vascular and stromal effects
important implications when dose-fractionation that are not seen with CFRT (Kirkpatrick and
schemes are compared for SABR. In addition, factors Dewhirst 2008; Fuks and Kolesnick 2005), normal
such as treatment delivery time and dose rate become tissue effects after SABR, preservation of immune
significant considerations for predicting cell kill dur- response (Lee et al. 2009; Curiel 2007), and the
ing SABR. For example, evaluation of clonogenic combination of effective mechanisms with targeted
survival in vitro when exposed to high doses of drugs.
radiation from 12 to 18 Gy shows that in glioma cell
lines, cell kill can be significantly affected by treat-
ment duration changing from 1.5 to 2 h (Benedict 3.2 Normal Tissue Radiobiology
et al. 1997). A review of this topic has concluded that and Tolerance
for treatments lasting longer than 30 min, significant
loss of cytotoxicity may be seen in high dose per With high dose per fraction treatments given with
fraction treatments (Fowler et al. 2004). More SABR, different types of normal tissue can possess
research is needed both in vitro and in vivo to better unique radiation tolerance characteristics. Serial
model high dose-fractional treatments (Timmerman structures such as airways, nerves, vessels, and
and Story 2006), including different radiobiologic bowel are linear or branching structures where
352 J. H. Heinzerling and R. D. Timmerman

including arterial oxygen pressure on room air (PO2)


and diffusing capacity for carbon monoxide (DLCO)
after high dose per fraction treatment (Timmerman
et al. 2003a, b). Decreases in lung capacity mea-
surements such as FEV1 and FVC are less commonly
seen. Because downstream effects are related to
upstream damage, targets in close proximity to large
airways such as those near the hilum and central chest
are especially prone to high levels of downstream
damage and should be treated with great care (see
section on central tumors for more discussion). Direct
damage to parallel functioning tissues such as the
alveoli occurs at relatively low threshold dose; how-
ever, the overall volume of parallel tissue in a healthy
organ is typically very large. Much of this parallel
functioning tissue constitutes a reserve (i.e., func-
Fig. 11 A plot of the logarithm of cell survival with increasing tional capacity beyond what is actually needed for
single fraction dose is initially curvilinear (known as the activities of daily life). So long as the reserve (extra
shoulder) followed by a linear relationship. Giving the same volume of tissue) is preserved, patients will avoid
total dose of radiation by multiple smaller fractions (e.g., 2 Gy) symptomatic dysfunction. Unlike serially functioning
repeats the shoulder with each fraction resulting in considerably
more cell survival than for a single large dose. The linear- tissues, delivering damaging dose above the threshold
quadratic formalism appropriately models the curvilinear creates little additional dysfunction within parallel
shoulder portion of the survival curve but overestimates cell tissue; rather, the key to avoiding toxicity is to min-
loss in the linear portion imize exposing volumes of tissue at or above the
threshold dose. As SABR is inherently a volume-
sparing technique, parallel tissue dysfunction can be
damage at any point along their path can cause dramatically minimized compared to conventional,
downstream dysfunction. This is in contrast to par- large volume irradiation techniques. For instance,
allel structures such as lung alveoli, kidney neph- rates of pneumonitis seen in SABR for lung tumors
rons, and gland acini, where damage to one portion are far less than CFRT (Timmerman et al. 2003a, b).
does not necessarily affect adjacent tissue. Within Therefore, as discussed above, the dose fall-off region
the thorax, both serial and parallel normal tissues or ‘‘gradient region’’ should be mechanistically
exist in close proximity including serially reduced to ensure that damage to parallel structures is
functioning small and large airways, esophagus, and kept to a minimum.
brachial plexus adjacent to parallel functioning Other serial tissues can also show dramatic
alveoli/capillary complexes. In addition, the heart, differences in toxicity between what is classically
pericardium, pleura, bones, and chest wall, which are seen in CFRT versus SABR. Esophagitis, while
difficult to assign as parallel versus serial have commonly seen with CFRT, is typically self limiting
unique mechanisms of injury and tolerance to high and resolves 2–4 weeks after treatment completion.
dose per fraction treatment. After SABR, however, high dose to the serial
In contrast to CFRT, which often causes minor, esophageal tissue can cause esophageal strictures.
repairable irritation of serially functioning airways, Pleural and pericardial effusions can also develop
SABR dose schedules can cause significant damage to with SABR dose schedules due to irritation of the
large and small airways within the lung leading to pericardium or pleura, especially in tumors adjacent
mucosal injury and downstream collapse. Similar to the heart or chest wall. Typically, these fluid col-
damage can be inflicted on blood vessels traveling lections will reabsorb without any necessary inter-
along the routes of these airways. Loss of lung vention, but they are important to characterize
parenchymal function (either ventilation or perfusion) because of their absence in CFRT. Other, more rare
most often leads to effects on oxygenation parameters late effects with SABR that have been described in
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 353

serial tissues include aneurysms, fistulas, and neur- Based on data suggesting a dose–response relation-
opathies, and should caution to investigators to ship in these patients, use of oligofractioned SABR
monitor doses to all serial functioning tissues was first explored in the inoperable patient
including large blood vessels and large nerves such as population.
brachial plexus, phrenic, and intercostal nerves. More Early retrospective experience using SABR for
follow up is still needed from previously completed primary lung tumors showed effective responses for
clinical trials to describe other late effects of SABR primary lung tumors with a wide variety of dose-
that have not yet manifested themselves. fractionation schemes (Blomgren et al. 1995; Nyman
et al. 2006; Uematsu et al. 2001; Uematsu et al.
1998). Yet, many of the criteria for patients to be
4 Clincal Results in Primary Lung treated differed, and most of the experiences had
Cancer small patient numbers treated with quite variable
dose–fractionation schemes. In addition, patients
SABR provides a local, ablative dose particularly were typically treated at one center, and often the
effective at eradicating gross visible tumor, making it follow up was short, making reporting of late toxici-
an ideal treatment for limited visible disease without ties seen with these treatments inadequate. Conse-
regional or distant spread. In contrast, SABR is not quently, to truly study SABR within an interpretable
particularly appropriate for treating microscopic dis- forum, investigation with clear, predefined selection
ease either adjuvantly or prophylactically because of criteria, consistent doses, strict quality assurance, and
its likelihood of causing collateral damage to normal adequate follow up is needed through the use of
tissues with high dose per fraction treatment. A dis- prospective trials.
ease that is localized after ideal staging workup and Indiana University carried out a phase I dose-
has a low probability of regional or distant metastatic escalation study in patients with stage I medically
spread represents the ideal setting for which the inoperable NSCLC to both evaluate toxicity for
principles of SABR could be applied and provide SABR in primary lung cancer and to determine the
benefit when compared to CFRT. Thus, small-cell maximum tolerated dose (MTD) (Timmerman et al.
lung cancer and advanced (node positive) non-small- 2003a, b; McGarry et al. 2005). Forty-seven patients
cell lung cancer (NSCLC) are conditions where with T1-2 N0 NSCLC were treated with consistent
SABR is unlikely to be used effectively except pos- SABR techniques in 3 fractions. Independent escala-
sibly as a boost to gross disease. Early stage NSCLC tion trials were performed for 3 different tumor
and limited lung metastases in patients with con- cohorts: T1, T2 \ 5 and T2 5–7 cm. All intrathoracic
trolled systemic disease are diseases where the prin- tumor locations were treated including central tumors.
ciples of SABR could be exploited to achieve higher Doses were escalated from 24 Gy over 3 fractions
rates of tumor control. (8 Gy per fraction) up to 72 Gy in 3 fractions (24 Gy
per fraction). The MTD was not reached for T1 or T2
tumors \ 5 cm despite reaching doses of 60–66 Gy
4.1 Medically Inoperable Patients in 3 fractions. For T2 tumors larger than 5 cm, the
with Early Stage Lung Cancer MTD was determined to be 66 Gy in 3 fractions after
dose limiting toxicity that included pneumonia and
Surgical resection remains standard therapy for pericardial effusion was seen at the 72 Gy dose level.
patients with stage I non-small-cell lung cancer Impressive tumor response was seen at all dose levels
(NSCLC), with 5 year survival rates of approximately (Fig. 12), but on longer follow up, ten local failures
60–70% (Naruke et al. 1988; Nesbitt et al. 1995). were seen out of 47 patients at a median follow up of
Patients determined to be medically inoperable have 15.2 months. Nine of the ten failures were in patients
been treated in the past with standard fractionated who received B16 Gy per fraction. As patients con-
radiotherapy, typically given to a dose of approxi- tinued to be followed, radiologic changes surrounding
mately 45–66 Gy in fractions of 1.8–2 Gy over the tumor were seen including fibrotic changes in the
6 weeks, with 5 year survival of approximately 10– lung (Matsuo et al. 2007), but were not typically
30% (Kaskowitz et al. 1993; Wisnivesky et al. 2005). associated with any symptoms. Often, these changes
354 J. H. Heinzerling and R. D. Timmerman

Fig. 12 Example of response seen after SABR in early stage primary lung cancer. Dramatic tumor response can be seen as early
as 2–3 months with total radiologic disappearance often by 1 year

can be mistaken for tumor recurrence (Matsuo et al. (Fakiris et al. 2009). Several series from the U.S.,
2007; Takeda et al. 2008). but follow up PET scan Europe, and Asia have duplicated local control rates
and biopsies showed no evidence of tumor recurrence seen in these clinical trials from Indiana, with varying
in the majority of patients treated in the higher dose dose and fractionation schemes. Local control rates
cohorts. range from 80 to 95% and are summarized in Table 1.
Because of the encouraging tumor response and Based on the encouraging results seen in the
tolerable toxicity seen in the phase I experience, the Indiana experience, the Radiation Therapy Oncology
Indiana group further evaluated SABR in this patient Group initiated a multi-institutional phase II study in
population with a 70 patient phase II trial (Timmer- 2002. RTOG 0236 completed accrual of 59 patients in
man et al. 2006; Fakiris et al. 2009). Small tumors October of 2006. Eligible patients were purely med-
were treated with 60 Gy in 3 fractions while larger ically inoperable (not confounded by those refusing
tumors received 66 Gy in 3 fractions, with 35 patients surgery) and included peripheral T1 or T2/3 tumors
enrolled to each cohort. The study was powered for a B5 cm. Patients with tumors within 2 cm of the
target local control rate of 80%, which would repre- ‘‘proximal bronchial tree’’ (Fig. 13) were excluded
sent a dramatic improvement over results seen with based on the toxicity seen in the phase II study from
CFRT. Patients continued to be followed for toxicity Indiana University.
related to treatment. The initial results of the trial Patients were treated to 60 Gy in 3 fractions based
were published early because of new detection of on planning without tissue heterogeneity correction
treatment related toxicity not seen in the phase I trial. (assuming the body is solid water). Forty-four patients
Actuarial 2 year local control was 95% with a median had T1 tumors, 11 had T2 tumors, and no patients
follow up of 17.5 months, and overall survival was enrolled had T3 tumors. Extensive central accredita-
56% at 2 years. The majority of deaths were found to tion, conduct, and dosimetry constraints were devel-
be related to comorbid illnesses seen in this frail oped prior to the opening of the trial by the RTOG
patient population, rather than death associated with Lung, Physics, and Image-Guided Therapy commit-
lung cancer. Again, toxicity was tolerable, with less tees to ensure meaningful quality assurance of SABR
than 20% of patients experiencing high grade toxicity, techniques and that patients received consistent
but there were 6 treatment related deaths. Severe treatment according to protocol guidelines at all of the
toxicity (Grades 3–5) was significantly more likely to participating centers. The results of this trial were
occur in patients with ‘‘central’’ tumors, defined as recently published in the 2010 theme issue on cancer
tumors near the proximal bronchial tree (see Sect. 5). for the Journal of the American Medical Association
The trial was recently updated after a median (Timmerman et al. 2010a, b). Severe toxicity was
follow up of 50 months showing 3-year local con- limited in this trial, with only 12.7% of patients with
trol and survival of 88 and 42%, respectively grade 3 treatment related toxicity, and only 3.6% with
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 355

Table 1 Summary of primary tumor control results for SABR treatment of stage I lung tumors
Author Treatment Local control Single Fraction Equivalent Dose (Dq = 2 Gy)
(%) (Timmerman et al. 2007a, b)
Europe/North America
Timmerman, 2003 (Timmerman et al. 8–16 Gy 9 3 60 (2 years) 20–44 Gy
2003a, b) McCarty et al. 2005) 18–24 Gy 9 3 90 (2 years) 50–68 Gy
Zimmerman, 2005 (Zimmermann et al. 12.5 Gy 9 3 87 (3 years) 43.5 Gy
2005)
Nyman, 2006 (Nyman et al. 2006) 15 Gy 9 3 80 (crude) 41 Gy
Timmerman, 2006 (Timmerman et al. 20–22 Gy 9 3 95 56–62 Gy
2006) (2 ? years)
Baumann, 2006 (Baumann et al. 2006) 15 Gy 9 3 80 (3 years) 41 Gy
Fritz, 2008 (Fritz et al. 2008) 30 Gy 9 1 81 (3 years) 30 Gy
Hof, 2007 (Hof et al. 2007) 19–24 Gy 9 1 50 (2 years) 19–24 Gy
26–30 Gy 9 1 72 (2 years) 26–30 Gy
Pennathur, 2007 (Pennathur et al. 2007) 20 Gy 9 1 84 (crude) 20 Gy
Asia
Nagata, 2005 (Nagata et al. 2005) 12 Gy 9 4 94 (3 years) 42 Gy
Xia, 2006 (Xia et al. 2006) 5 Gy 9 10 95 (3 years) 32 Gy
Hara, 2006 (Hara et al. 2006) 30–34 Gy 9 1 80 (3 years) 30–34 Gy
Koto, 2007 (Koto et al. 2007) 15 Gy 9 3 or T1 78, T2 40 41–46 Gy
7.5 Gy 9 6 (3-years)

grade 4 toxicity. Only one failure was seen at the have published prospective results of SABR where
primary tumor site, leading to a 3-year primary tumor treatment was based on patient characteristics related
control rate of 98%. The local control rate was to tumor size and location (Lagerwaard et al. 2008).
determined to be 91% with three additional patients After treating 200 ? patients (80% were medically
having failure within the involved lobe outside of the inoperable), local control was over 90% while severe
treated area. Regional failure within hilar or medias- late toxicity was under 5% with their ‘‘risk adapted’’
tinal lymph nodes was low despite non-surgical approach. A Nordic study group similarly reported
staging, with a 3-year loco-regional control of 87%. 65% 2-year overall survival in a group of 57 medi-
Eleven patients, however, failed in distant sites, the cally inoperable patients. Local control at 2-years was
majority within 1 year of treatment. Despite these 93% using their 3 fraction SABR regimen (Baumann
failures, disease-free and overall survival were also et al. 2009). The Japanese Clinical Oncology Group
encouraging in this trial with 3-year rates of 48 and has been accruing to a prospective trial treating
56%, respectively. Because these survival results are medically inoperable patients with T1 tumors using 4
only modestly poorer than the results with definitive fractions of 12 Gy. This trial continues to accrue, but
surgical resection despite the frail population in which the results of utilizing this fractionation scheme in
the study was conducted (Naruke et al. 1988; Nesbitt medically operable patients was recently presented
et al. 1995; Ginsberg and Rubinstein 1995), explora- and are discussed below.
tion of utilizing SABR techniques within the medi- The ideal dose for SABR treatment of early stage
cally operable population was deemed feasible and is lung tumors continues to be explored in the clinical
discussed below. trial setting. Dose-fractionation schemes differ from
Other multicenter prospective trials utilizing reported trials as shown in Table 1, and the follow-up
SABR in the medically inoperable population have time for these existing series is still limited. The
been performed internationally. The Dutch Group optimal dose-fractionation schedule likely varies with
356 J. H. Heinzerling and R. D. Timmerman

Fig. 13 Diagram showing the anatomical delineation of ‘‘central or perihilar’’ tumors

tumor stage and location, and no randomized studies used in RTOG 0236 in a randomized phase III trial to
have been completed thus far comparing different follow.
fractionation schemes. From the experience from
Indiana, severe toxicity was high in tumors within the
central lung (Fig. 12), leading to exclusion of these 4.2 Medically Operable Patients
patients on the subsequent RTOG 0236 trial. The
RTOG has now opened a seamless phase I/II trial to Because of the high tumor control and acceptable
address the question of escalating doses in 5 fractions toxicity seen with SABR for early stage NSCLC in the
for centrally located tumors (RTOG 0813). Patients inoperable population, the use of SABR in patients
will start at a dose of 50 Gy in 5 fractions with able to undergo surgical resection is being explored in
escalation up to 12 Gy per fraction to determine the the U.S., Europe, and Asia. A large retrospective
MTD for central tumors in the inoperable patient experience from Japan included a significant number
population. The RTOG has also begun to explore of operable patients and showed a 3-year survival of
other dose-fractionation schemes in peripherally 88% (Onishi et al. 2004). On the basis of this data, the
located tumors, comparing 34 Gy in 1 fraction to Japan Clinical Oncology Group initiated a clinical trial
48 Gy in 4 fractions. The least toxic of these two evaluating SABR in peripheral T1 NSCLC for oper-
regimens will be compared to the 20 Gy 9 3 regimen able patients (JCOG 0403). All patients were treated
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 357

with a dose of 48 Gy in 4 fractions. The initial results implemented. As discussed in Sect. 4, normal tissue
of this trial were recently presented at the American toxicity seen with SABR can often involve severe
Society of Therapeutic Radiology and Oncology destruction of medium and small airway passages
annual meeting in San Diego in November 2010. 3- with consequential late effects including airway
year overall survival was 76%, an encouraging result strictures leading to downstream dysfunction. While
for the first prospective multicenter trial in this pop- normal tissue dose constraints are implemented into
ulation. However, there were 25 patients with pro- prospective trials, these constraints are often based on
gressive disease, and the 3-year local progression free a small number of cases where the toxicity were
survival was only 68.5%, which is significantly lower observed, mathematical models, or even educated
than previous experience in the inoperable population guessing because of the short follow-up experience
suggesting that there may be a need for higher doses in within the literature. Observations of toxicity continue
these patients. to be reported and analyzed in a prospective fashion
In the U.S, the RTOG has completed a trial (RTOG and linked with dosimetric endpoints in order to
0618) evaluating SABR treatment of early stage modify SABR dose constraints for normal tissues.
NSCLC in the operable population to evaluate SABR Some of the main toxicities observed in the pro-
as an alternative to surgery in these patients. Eligible spective experience of SABR for lung tumors will be
patients included peripheral T1 or T2/3 tumors discussed here.
B5 cm. Patients were all considered to be reasonable The phase I study from Indiana established the
candidates for surgical resection by a qualified tho- maximum tolerated dose for 3 fraction SABR in the
racic surgeon and baseline pulmonary function tests. medically inoperable population (Timmerman et al.
Because these patients have a high probability of 2003a, b). All patients underwent pre and post-treat-
cure, strict follow up for response was performed in ment evaluation of pulmonary function and were
order to promptly identify local failures so that sal- closely monitored for a minimum observation period
vage therapy with surgical resection could be per- for toxicity prior to dose escalation. Common low
formed. Based on the surgical literature from the U.S., grade toxicity included fatigue in all patients. Car-
SABR in this population will be required to achieve a diopulmonary toxicity was reasonable and included
local control rate of 90% or better to compete with 10 patients who had a 10% decline in one measured
surgical resection. This trial has now completed and value of pulmonary function (FEV1, FVC, DLCO,
focus within North American cooperative trial groups and PO2). The majority of these patients had eventual
has turned to direct randomized comparison between return to baseline. The two parameters that most
surgical resection and SABR for early stage lung commonly declined after treatment were DLCO and
cancer. The RTOG and American College of Sur- PO2. Two patients had a dose limiting toxicity,
geons Oncology Group (ACOSOG) have opened a including one patient with grade 3 pneumonitis
randomized phase III trial comparing sublobar (despite this being predicted as the most likely tox-
resection to SABR in high risk operable patients with icity), and one with grade 3 hypoxemia. Given the
stage I NSCLC. With a primary endpoint of 3-year very frail patient population in this trial, the toxicity
overall survival, the trial explores whether SABR can observed was quite acceptable.
be an acceptable alternative to surgical resection in As discussed above, the phase II study from the
this patient population with an overall survival rate Indiana group exposed higher levels of severe pul-
not more than 10% less than patients receiving sub- monary toxicity in tumors adjacent to the central and
lobar resection. perihilar structures within the lung. Low grade tox-
icities in this trial included fatigue, musculoskeletal
pain, and radiation pneumonitis. Fourteen patients
5 Toxicity had more severe toxicity, including decline in pul-
monary function tests, pleural effusions, and pneu-
Because of the high doses per fraction that SABR monias, leading to 6 deaths that may have been a
utilizes, there is potential for higher rates of normal result of the treatment. The majority of deaths were
tissue toxicity than conventionally fractionated radi- due to pneumonia, and on multivariate analysis,
ation if the techniques discussed above are not central tumor location was a strong predictor of
358 J. H. Heinzerling and R. D. Timmerman

having a severe toxicity. Four of the six deaths were opposite is occurring within oncologic therapy. As
in patients with central tumors, and these patients had systemic therapy proves more effective, local failure
an 11-fold increased risk of having a severe toxicity is becoming an increasingly more common method of
compared to patients with peripheral tumors. As sta- failure, making local control progressively more
ted above, investigation into different SABR regimens critical to patient outcome. Thus, the techniques and
to treat tumors located within the zone of the proxi- ablative doses utilized with SABR will become more
mal bronchial tree is ongoing. important not only in early stage disease, but also in
Based on the phase II results from Indiana, RTOG metastatic disease as a measure for consolidation or
0236 limited 3 fraction SABR to peripheral tumors ablation of resistant cancer deposits after systemic
only, and subsequently showed improved rates of therapy. In addition, customization of therapy to
toxicity for treatment of medically inoperable patient specific tumor characteristics will become
patients. Grade 3–4 toxicity was seen in 15 patients increasingly important in the future as biological,
(28%) treated on RTOG 0236, and no treatment clinical, and technical research within oncology cre-
related deaths were seen. Most of the toxicity seen ates paradigms to facilitate adaptive therapy (Song
was pulmonary or musculoskeletal. Dose-volume et al. 2005; Martinez et al. 2001; Bortfeld and
information collected from this and other early Paganetti 2006). Within adaptive therapy, pretreat-
experience with SABR will be used to both validate ment diagnostic information including imaging,
and modify parameters used for normal tissue toxicity staging, and tissue characteristics (proteomic,
in these patients (Dunlap et al. 2010). genomics, and predictive assays) will be integrated to
design patient specific therapy (Potti et al. 2006).
Patients can be monitored during treatment with
6 Conclusions similar methods and treatments can be adjusted
including the need for adjuvant therapies and to avoid
SABR has utilized innovation within the engineering toxicity from treatment. This paradigm avoids the
and physics of radiation therapy to increase treatment ‘‘one size fits all’’ mantra in current oncologic ther-
accuracy and to allow delivery of oligofractionated, apy, and utilizes a tailored approach to constantly
ablative doses of radiation. This technological reevaluate and respond to queues in order to redirect
advancement has allowed the exploration of high dose therapy toward better patient outcome. As we con-
per fraction treatments leading to observation of tinue towards this goal of adapting therapy, it will
unique radiobiological outcomes that have challenged continue to be crucial to utilize well designed pro-
the principles of conventional fractionation. The spective trials so that therapeutic tools such as SABR
technologic and biologic benefits of SABR have been can be refined to their optimal potential.
observed most dramatically in patients with early
stage lung cancer. Use of SABR in these patients has
now been established as the standard treatment option
References
for medically inoperable patients through careful
study in the prospective, multi-institutional clinical
Balter JM, Wright JN, Newell LJ et al (2005) Accuracy of a
trials described above. Toxicity of this treatment has wireless localization system for radiotherapy. Int J Radiate
been well characterized in the clinical trials, allowing Oncol Biol Phys 61:933–937
for appropriate selection of candidates for SABR. Baumann P, Nyman J, Lax I et al (2006) Factors important for
Because of the encouraging control rates seen in efficacy of stereotactic body radiotherapy of medically
inoperable stage I lung cancer. A retrospective analysis of
medically inoperable patients, study of SABR has patients treated in the Nordic countries. Acta Oncol
now been extended to medically operable patients and 45:787–795
is being compared to surgical resection in ongoing Baumann P, Nyman J, Hoyer M et al (2009) Outcome in a
randomized clinical trials. prospective phase II trial of medically inoperable stage I
non-small-cell lung cancer patients treated with stereotactic
As systemic therapy becomes more effective for body radiotherapy. J Clin Oncol 27:3290–3296
solid tumors, it was thought that local treatments such Benedict SH, Lin PS, Zwicker RD, Huang DT, Schmidt-Ullrich
as radiotherapy and surgical resection would be less RK (1997) The biological effectiveness of intermittent
utilized in cancer therapy. Interestingly, however, the irradiation as a function of overall treatment time:
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 359

development of correction factors for linac-based stereo- cell lung cancer. Lung cancer study group. Ann Thorac Surg
tactic radiotherapy. Int J Radiate Oncol Biol Phys 37:765– 60:615–622 discussion 22–23
769 Guerrero M, Li XA (2004) Extending the linear-quadratic
Benedict SH, Yenice KM, Followill D et al (2010) Stereotactic model for large fraction doses pertinent to stereotactic
body radiation therapy: the report of AAPM task group 101. radiotherapy. Phys Med Biol 49:4825–4835
Med Phys 37:4078–4101 Hall EJ, Gross W, Dvorak RF, Kellerer AM, Rossi EH (1972)
Blomgren H, Lax I, Naslund I, Svanstrom R (1995) Stereotactic Survival curves and age response functions for Chinese
high dose fraction radiation therapy of extracranial tumors hamster cells exposed to x-rays or high LET alpha-particles.
using an accelerator. Clinical experience of the first thirty- Radiate Res 52:88–98
one patients. Acta Oncol 34:861–870 Hara R, Itami J, Kondo T et al (2006) Clinical outcomes of
Bortfeld T, Paganetti H (2006) The biologic relevance of daily single-fraction stereotactic radiation therapy of lung tumors.
dose variations in adaptive treatment planning. Int J Radiate Cancer 106:1347–1352
Oncol Biol Phys 65:899–906 Heinzerling JH, Anderson JF, Papiez L et al (2008) Four-
Cardinale RM, Wu Q, Benedict SH, Kavanagh BD, Bump E, dimensional computed tomography scan analysis of tumor
Mohan R (1999) Determining the optimal block margin on and organ motion at varying levels of abdominal compression
the planning target volume for extracranial stereotactic during stereotactic treatment of lung and liver. Int J Radiate
radiotherapy. Int J Radiate Oncol Biol Phys 45:515–520 Oncol Biol Phys 70:1571–1578
Chawla S, Chen Y, Katz AW et al (2009) Stereotactic body Herfarth KK, Debus J, Lohr F et al (2001) Stereotactic single-
radiotherapy for treatment of adrenal metastases. Int J dose radiation therapy of liver tumors: results of a phase I/II
Radiate Oncol Biol Phys 75:71–75 trial. J Clin Oncol 19:164–170
Chang SD, Main W, Martin DP, Gibbs IC, Heilbruon MP Hof H, Muenter M, Oetzel D, Hoess A, Debus J, Herfarth K
(2003) An analysis of the accuracy of the cyberknife: a (2007) Stereotactic single-dose radiotherapy (radiosurgery)
robotic frameless stereotactic radio surgical system. Neuro- of early stage non-small-cell lung cancer (NSCLC). Cancer
surgery 52:140–146 discussion 6–7 110:148–155
Chang EL, Shiu AS, Lii MF et al (2004) Phase I clinical Jaffray DA, Siewerdsen JH, Wong JW, Martinez AA (2002)
evaluation of near-simultaneous computed tomographic Flat-panel cone-beam computed tomography for image-
image-guided stereotactic body radiotherapy for spinal guided radiation therapy. Int J Radiate Oncol Biol Phys
metastases. Int J Radiate Oncol Biol Phys 59:1288–1294 53:1337–1349
Chen QS, Weinhous MS, Deibel FC, Ciezki JP, Macklis RM Kaskowitz L, Graham MV, Emami B, Halverson KJ, Rush C
(2001) Fluoroscopic study of tumor motion due to breath- (1993) Radiation therapy alone for stage I non-small cell
ing: facilitating precise radiation therapy for lung cancer lung cancer. Int J Radiate Oncol Biol Phys 27:517–523
patients. Med Phys 28:1850–1856 Kimura T, Hirokawa Y, Murakami Y et al (2004) Reproduc-
Chen GT, Kung JH, Beudette KP (2004) Artifacts in computed ibility of organ position using voluntary breath-hold method
tomography scanning of moving objects. Semin Radiate with spirometer for extracranial stereotactic radiotherapy.
Oncol 14:19–26 Int J Radiate Oncol Biol Phys 60:1307–1313
Curiel TJ (2007) Tregs and rethinking cancer immunotherapy. Kini VR, Vedam SS, Keall PJ, Patil S, Chen C, Mohan R
J Clin Invest 117:1167–1174 (2003) Patient training in respiratory-gated radiotherapy.
Dunlap NE, Cai J, Biedermann GB et al (2010) Chest wall Med Dosim 28:7–11
volume receiving[30 Gy predicts risk of severe pain and/or Kirkpatrick JP, Dewhirst MW (2008) Analytic solution to
rib fracture after lung stereotactic body radiotherapy. Int J steady-state radial diffusion of a substrate with first-order
Radiate Oncol Biol Phys 76:796–801 reaction kinetics in the tissue of a Krogh’s cylinder. Radiate
Fakiris AJ, McCarrty RC, Yiannoutsos CT et al (2009) Res 169:350–354
Stereotactic body radiation therapy for early-stage non- Kooch N, Liu HE, Starkschall G et al (2004) Evaluation of
small-cell lung carcinoma: four-year results of a prospec- internal lung motion for respiratory-gated radiotherapy
tive phase II study. Int J Radiate Oncol Biol Phys 75: using MRI: Part I—correlating internal lung motion with
677–682 skin fiducial motion. Int J Radiate Oncol Biol Phys
Fowler JF, Welsh JS, Howard SP (2004) Loss of biological 60:1459–1472
effect in prolonged fraction delivery. Int J Radiate Oncol Koong AC, Le QT, Ho A et al (2004) Phase I study of
Biol Phys 59:242–249 stereotactic radiosurgery in patients with locally advanced
Fritz P, Kraus HJ, Blaschke T et al (2008) Stereotactic, high pancreatic cancer. Int J Radiate Oncol Biol Phys
single-dose irradiation of stage I non-small cell lung cancer 58:1017–1021
(NSCLC) using four-dimensional CT scans for treatment Kuriyama K, Onishi H, Sano N et al (2003) A new irradiation
planning. Lung Cancer 60:193–199 unit constructed of self-moving gantry-CT and linac. Int J
Fuks Z, Kolesnick R (2005) Engaging the vascular component Radiate Oncol Biol Phys 55:428–435
of the tumor response. Cancer Cell 8:89–91 Koto M, Takai Y, Ogawa Y et al (2007) A phase II study on
Fuss M, Salter BJ, Cheek D, Sadeghi A, Hevesy JM, Herman stereotactic body radiotherapy for stage I non-small cell
TS (2004) Repositioning accuracy of a commercially lung cancer. Radiother Oncol 85:429–434
available thermoplastic mask system. Radiother Oncol 71: Lagerwaard FJ, Van Sornsen de Koste JR, Nijssen-Visser MR
339–345 et al (2001) Multiple ‘‘slow’’ CT scans for incorporating
Ginsberg RJ, Rubinstein LV (1995) Randomized trial of lung tumor mobility in radiotherapy planning. Int J Radiate
lobectomy versus limited resection for T1 N0 non-small Oncol Biol Phys 51:932–937
360 J. H. Heinzerling and R. D. Timmerman

Lagerwaard FJ, Haasbeek CJ, Smit EF, Slotman BJ, Senan S stereotactic body frame. Int J Radiate Oncol Biol Phys
(2008) Outcomes of risk-adapted fractionated stereotactic 63:1427–1431
radiotherapy for stage I non-small-cell lung cancer. Int J Naruke T, Goya T, Tsuchiya R, Suemasu K (1988) Prognosis
Radiate Oncol Biol Phys 70:685–692 and survival in resected lung carcinoma based on the new
Lax I, Blomgren H, Naslund I, Svanstorm R (1994) Stereotactic international staging system. J Thorax Cardiovascular Surge
radiotherapy of malignancies in the abdomen. Methodolo- 96:440–447
gical aspects. Acta Oncol 33:677–683 Negoro Y, Nagata Y, Aoki T et al (2001) The effectiveness of
Lee Y, Auh SL, Wang Y et al (2009) Therapeutic effects of an immobilization device in conformal radiotherapy for
ablative radiation on local tumor require CD8 ? T cells: lung tumor: reduction of respiratory tumor movement and
changing strategies for cancer treatment. Blood 114:589–595 evaluation of the daily setup accuracy. Int J Radiate Oncol
Leskell L (1951) The stereotactic method and radiosurgery of Biol Phys 50:889–898
the brain. Acta Chir Scand 102:316–319 Nesbitt J, Putnam JB Jr, Walsh GL, Roth JA, Mountain CF
Liu R, Buatti JM, Howes TL, Dill J, Modrick JM, Meeks SL (1995) Survival in early-stage non-small cell lung cancer.
(2006) Optimal number of beams for stereotactic body Ann Thorax Surge 60:466–472
radiotherapy of lung and liver lesions. Int J Radiate Oncol Nyman J, Johansson KA, Hulten U (2006) Stereotactic hypo
Biol Phys 66:906–912 fractionated radiotherapy for stage I non-small cell lung
Liu HH, Balter P, Tutt T et al (2007) Assessing respiration- cancer—mature results for medically inoperable patients.
induced tumor motion and internal target volume using Lung Cancer 51:97–103
four-dimensional computed tomography for radiotherapy of O’Dell WG, Schell MC, Reynolds D, Okunieff R (2002) Dose
lung cancer. Int J Radiate Oncol Biol Phys 68:531–540 broadening due to target position variability during frac-
Loo BW, Chang JY, Dawson LA et al (2011) Stereotactic tionated breath-held radiation therapy. Med Phys
ablative radiotherapy: what’s in a name? Pract Radiate 29:1430–1437
Oncol 1:38–39 Onishi H, Araki T, Shirato H et al (2004) Stereotactic hypo
Madsen BL, Hsi RA, Pham HT, Fowler JR, Esagui L, Corman J fractionated high-dose irradiation for stage I nonsmall cell
(2007) Stereotactic hypofractionated accurate radiotherapy lung carcinoma: clinical outcomes in 245 subjects in a
of the prostate (SHARP), 33.5 Gy in five fractions for Japanese multiinstitutional study. Cancer 101:1623–1631
localized disease: first clinical trial results. Int J Radiate Papiez L, Timmerman R, DesRosiers C, Randall M (2003)
Oncol Biol Phys 67:1099–1105 Extracranial stereotactic radioablation: physical principles.
Mageras GS, Yorke E (2004) Deep inspiration breath hold and Acta Oncol 42:882–894
respiratory gating strategies for reducing organ motion in Park C, Papiez L, Zhang S, Story M, Timmerman RD (2008)
radiation treatment. Semin Radiate Oncol 14:65–75 Universal survival curve and single fraction equivalent
Mageras GS, Pevsner A, Yorke ED et al (2004) Measurement dose: useful tools in understanding potency of ablative
of lung tumor motion using respiration-correlated CT. Int J radiotherapy. Int J Radiate Oncol Biol Phys 70:847–852
Radiate Oncol Biol Phys 60:933–941 Pennathur A, Luketich JD, Burton S et al (2007) Stereotactic
Marks LB (1995) Extrapolating hypofractionated radiation radiosurgery for the treatment of lung neoplasm: initial
schemes from radiosurgery data: regarding Hall et al. experience. Ann Thorac Surg 83:1820–1824 discussion
IJROBP 21:819–824; 1991 and Hall and Brenner (1993) 4–5
IJROBP 25:381–385. Int J Radiat Oncol Biol Phys 1995; Potters L, Steinberg M, Rose C et al (2004) American Society
32:274–276 for Therapeutic Radiology and Oncology and American
Martinez AA, Yan D, Lockman D et al (2001) Improvement in College of Radiology practice guideline for the performance
dose escalation using the process of adaptive radiotherapy of stereotactic body radiation therapy. Int J Radiate Oncol
combined with three-dimensional conformal or intensity- Biol Phys 60:1026–1032
modulated beams for prostate cancer. Int J Radiate Oncol Potti A, Mukherjee S, Petersen R et al (2006) A genomic
Biol Phys 50:1226–1234 strategy to refine prognosis in early-stage non-small-cell
Matsuo Y, Nagata Y, Mizowaki T et al (2007) Evaluation of lung cancer. N Engl J Med 355:570–580
mass-like consolidation after stereotactic body radiation Rossi HH, Kellerer AM (1972) Radiation carcinogenesis at low
therapy for lung tumors. Int J Clin Oncol 12:356–362 doses. Science 175:200–202
McGarry RC, Papiez L, Williams M, Whit ford T, Timmerman Rusthoven KE, Kavanagh BD, Burri SH et al (2009a) Multi-
RD (2005) Stereotactic body radiation therapy of early- institutional phase I/II trial of stereotactic body radiation
stage non-small-cell lung carcinoma: phase I study. Int J therapy for lung metastases. J Clin Oncol 27:1579–1584
Radiate Oncol Biol Phys 63:1010–1015 Rusthoven KE, Kavanagh BD, Cardenes H et al (2009b) Multi-
Murphy MJ (1997) An automatic six-degree-of-freedom image institutional phase I/II trial of stereotactic body radiation
registration algorithm for image-guided frameless stereo- therapy for liver metastases. J Clin Oncol 27:1572–1578
taxic radiosurgery. Med Phys 24:857–866 Schweikard A, Shiomi H, Adler J (2004) Respiration tracking
Murphy MJ, Martin D, Whyte R, Hai J, Ozhasoglu C, Le QT in radiosurgery. Med Phys 31:2738–2741
(2002) The effectiveness of breath-holding to stabilize lung Sharp GC, Jiang SB, Shimizu S, Shirato H (2004) Prediction of
and pancreas tumors during radiosurgery. Int J Radiate respiratory tumour motion for real-time image-guided
Oncol Biol Phys 53:475–482 radiotherapy. Phys Med Biol 49:425–440
Nagata Y, Takayama K, Matsuo Y et al (2005) Clinical Shirato H, Shimizu S, Shimizu T, Nishioka T, Miyasaka K
outcomes of a phase I/II study of 48 Gy of stereotactic body (1999) Real-time tumour-tracking radiotherapy. Lancet
radiotherapy in 4 fractions for primary lung cancer using a 353:1331–1332
Stereotactic Ablative Radiotherapy for Early Stage Lung Cancer 361

Shirato H, Shimizu S, Kunieda T et al (2000) Physical aspects stage I non-small cell lung cancer: a 5-year experience. Int J
of a real-time tumor-tracking system for gated radiotherapy. Radiate Oncol Biol Phys 51:666–670
Int J Radiate Oncol Biol Phys 48:1187–1195 Uematsu M, Shioda A, Tahara K et al (1998) Focal, high dose,
Sonke JJ, Zijp L, Remeijer P, van Herk M (2005) Respiratory and fractionated modified stereotactic radiation therapy for
correlated cone beam CT. Med Phys 32:1176–1186 lung carcinoma patients: a preliminary experience. Cancer
Song W, Schaly B, Bauman G, Battista J, Van Dyk J (2005) 82:1062–1070
Image-guided adaptive radiation therapy (IGART): radio- Vedam SS, Keall PJ, Kini VR, Mohan R (2001) Determining
biological and dose escalation considerations for localized parameters for respiration-gated radiotherapy. Med Phys
carcinoma of the prostate. Med Phys 32:2193–2203 28:2139–2146
Stevens CWM, Munden RF, Forster KM et al (2001) Respi- Verrellen D, Soete G, Linthout N et al (2003) Quality assurance
ratory-driven lung tumor motion is independent of tumor of a system for improved target localization and patient set-
size, tumor location, and pulmonary function. Int J Radiate up that combines real-time infrared tracking and stereo-
Oncol Biol Phys 51:62–68 scopic x-ray imaging. Radiothermy Oncol 67:129–141
Takeda A, Kunieda E, Takeda T et al (2008) Possible Wang LT, Solberg TD, Medin PM, Boone R (2001) Infrared
misinterpretation of demarcated solid patterns of radiation patient positioning for stereotactic radiosurgery of extra-
fibrosis on CT scans as tumor recurrence in patients cranial tumors. Comput Biol Med 31:101–111
receiving hypofractionated stereotactic radiotherapy for Wang L, Fienberg S, Chen L, Pasklev K, Ma CC (2006) Benefit
lung cancer. Int J Radiate Oncol Biol Phys 70:1057–1065 of three-dimensional image-guided stereotactic localization
Timmerman RD, Story M (2006) Stereotactic body radiation in the hypofractionated treatment of lung cancer. Int J
therapy: a treatment in need of basic biological research. Radiate Oncol Biol Phys 66:738–747
Cancer J 12:19–20 Wang Z, Wu QJ, Marks LB, Larrier N, Yin FF (2007) Cone-
Timmerman R, Papiez L, Suntharalingam M (2003a) Extra- beam CT localization of internal target volumes for
cranial stereotactic radiation delivery: expansion of tech- stereotactic body radiotherapy of lung lesions. Int J Radiate
nology beyond the brain. Techno Cancer Res Treat Oncol Biol Phys 69:1618–1624
2:153–160 Wisnivesky JP, Bonomi M, Henschke C, Iannuzzi M, McGinn
Timmerman R, Papiez L, McGarry R et al (2003b) Extracranial T (2005) Radiation therapy for the treatment of unresected
stereotactic radio ablation: results of a phase I study in stage I-II non-small cell lung cancer. Chest 128:1461–1467
medically inoperable stage I non-small cell lung cancer. Wulf J, Hadinger U, Oppitz U, Olshausen B, Flentje M (2000)
Chest 124:1946–1955 Stereotactic radiotherapy of extracranial targets: CT-simu-
Timmerman R, McCarty R, Yiannoutsos C et al (2006) Excessive lation and accuracy of treatment in the stereotactic body
toxicity when treating central tumors in a phase II study of frame. Radiothermy Oncol 57:225–236
stereotactic body radiation therapy for medically inoperable Xia T, Li H, Sun Q et al (2006) Promising clinical outcome of
early-stage lung cancer. J Clin Oncol 24:4833–4839 stereotactic body radiation therapy for patients with inop-
Timmerman RD, Kavanagh BD, Cho LC, Papiez L, Xing L erable Stage I/II non-small-cell lung cancer. Int J Radiate
(2007a) Stereotactic body radiation therapy in multiple Oncol Biol Phys 66:117–125
organ sites. J Clin Oncol 25:947–952 Yenice KM, Lovelock DM, Hunt MA et al (2003) CT image-
Timmerman RD, Park C, Kavanagh BD (2007b) The North guided intensity-modulated therapy for paraspinal tumors
American experience with stereotactic body radiation using stereotactic immobilization. Int J Radiate Oncol Biol
therapy in non-small cell lung cancer. J Thorac Oncol Phys 55:583–593
2:S101–S112 Yin F, Kim JG, Haughton C et al (2001) Extracranial
Timmerman R, Paulus R, Galvin J (2010a) Stereotactic body radiosurgery: immobilizing liver motion in dogs using
radiation therapy for inoperable early stage lung cancer. high-frequency jet ventilation and total intravenous anes-
JAMA 303(11):1170–1176 thesia. Int J Radiate Oncol Biol Phys 49:211–216
Timmerman R, Paulus R, Galvin J et al (2010b) Stereotactic Zimmermann FB, Geinitz H, Schill S et al (2005) Stereotactic
body radiation therapy for inoperable early stage lung hypofractionated radiation therapy for stage I non-small cell
cancer. JAMA 303:1070–1076 lung cancer. Lung Cancer 48:107–114
Uematsu M, Shioda A, Suda A et al (2001) Computed
tomography-guided frameless stereotactic radiotherapy for
Postoperative Radiotherapy for Non-Small
Cell Carcinoma
Ellen Kim and Mitchell Machtay

Contents Abstract
The role of post-operative radiotherapy (PORT)
1 Introduction.............................................................. 363 in treatment of non-small cell lung cancer
2 Patterns of Failure................................................... 364 (NSCLC) is unclear. Currently, the available
evidence suggests that PORT is indicated for
3 Results of PORT in Stage I NSCLC ..................... 364
stage II and III (node-positive) NSCLC, but not
4 Results of PORT in Stage II and III for stage I NSCLC. However, it is still disputed
(Node-Positive) NSCLC .......................................... 365
in which cases patterns of failure necessitate
5 Indications for PORT.............................................. 366 further treatment following surgery, and if it is
6 Toxicity and Mortality Risks of PORT ................ 367 warranted, the question becomes whether to do
radiotherapy instead of or with (before, during, or
7 Techniques for PORT ............................................. 368
after) chemotherapy. Study of PORT is further
8 PORT and Adjuvant Chemotherapy .................... 368 complicated by the improvement of radiotherapy
9 Conclusions and Future Directions ....................... 369 techniques in recent decades, and the shortage of
References.......................................................................... 369 well-defined prospective randomized trials since
their implementation. Advances in technology
have allowed more precise administration of
higher doses of radiation with lower exposure of
healthy tissues, resulting in greater local control
and lower treatment-related toxicities. Weighing
the benefits and risks to determine the overall
usefulness of PORT is difficult and will require
future studies.

1 Introduction

M. Machtay (&) Lung cancer treatment and prognosis depends on


Department of Radiation Oncology, several factors, including tumor histology, patient
Case Western Reserve University, performance status and extent of disease. The latter is
Cleveland, OH, USA
e-mail: mitchell.machtay@uhhospitals.org usually indicated by TNM stage. Clinical staging is
relatively unreliable, with disease often restaged to a
E. Kim
Case Western Reserve University School of Medicine, more advanced stage according to results of patho-
Cleveland, OH, USA logic staging following surgery.

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 363
DOI: 10.1007/174_2011_315, Ó Springer-Verlag Berlin Heidelberg 2011
364 E. Kim and M. Machtay

Although it has not been rigorously proven through 1987 and 1993, the largest study evaluating PORT in
randomized trials, surgery is currently considered the N2 NSCLC at the time. The patients were divided
standard primary treatment for medically operable into a group who received surgery alone and a group
patients with non-small cell lung cancer (NSCLC) who received surgery and thoracic PORT; the groups
who do not have evidence of mediastinal or distant were otherwise similar in gender, age, histology,
tumor involvement. Surgery optimizes the chance for number of lymph nodes involved, etc. The actuarial
local control and provides maximal pathologic stag- 4-year local recurrence rate and actuarial 4-year sur-
ing information to guide future therapy. vival rate were 60/17% for surgery alone and 22/43%
Adjuvant chemotherapy is now recommended for for surgery plus PORT. These results were highly
selected patients with NSCLC resected stage II–III. This statistically significantly in favor of PORT, and the
is based on several well-designed, ‘‘positive’’ random- authors concluded that adjuvant thoracic radiotherapy
ized trials. Postoperative radiation therapy (PORT) for may improve local control and survival.
NSCLC, in contrast, remains controversial. PORT Adjuvant chemotherapy, once controversial like
improves local control, particularly for node-positive PORT, has now been established as beneficial based
(stage II/III disease), but its effect on survival is unclear. on randomized clinical trials (Arriagada et al. 2004;
Historical prospective randomized trials to study PORT Strauss et al. 2008; Winton et al. 2005; NSCLC meta-
outcomes mostly came from the 1960 and 1970s, prior to analysis collaborative group 2010). The benefit of
major advances in radiation techniques. These older chemotherapy appears to be related to a slight (4–15%)
studies not only failed to show a survival benefit to decrease in the rate of distant metastases. However,
PORT, but concluded a detrimental effect in stage I and II adjuvant chemotherapy alone does not seem sufficient
disease. However, recent studies suggest that PORT can to provide adequate loco regional control. A retro-
improve not only local control but also actual survival for spective study of 98 patients by Taylor et al. (2003)
pathologic N2 patients. Patients with pathologically found stage IIB and IIIA NSCLC patients who
resected N2/IIIA disease should be strongly considered received surgery and adjuvant chemotherapy had
for PORT in addition to adjuvant chemotherapy. 5-year actuarial local control rate of only 54%. A recent
study by Le Pechoux (2011) likewise found a rate of
local–regional failure in excess of 20% following
2 Patterns of Failure complete resection and adjuvant chemotherapy of
pathological stage II–III N2 NSCLC, confirming the
In order to assess the potential usefulness of adjuvant need to continue investigating the potential advantages
therapies, it is essential to understand the patterns of of additional PORT.
failure after surgery. However, as shown in a multi-
institutional retrospective study of 306 patients with
resected stage I NSCLC, the definitions of patterns of 3 Results of PORT in Stage I NSCLC
failure can have a dramatic impact on the reported data
on patterns of failure (Varlotto et al. 2010). This may Most of the prospective studies of outcomes of PORT
contribute to the wide range, 20–50%, of reported for early stage NSCLC were performed prior to 1985.
rates of local–regional recurrence, particularly in the This was before the routine use of computed tomog-
1980 and 1990s (Kelsey et al. 2006). Varlotto and raphy (CT) and positron emission tomography (PET)
Colleagues, in this series, reported a 29% local failure scans. It was also prior to the widespread use of
rate for stage I NSCLC when using a definition whereby modern linear accelerators and 3-dimensional (3D)
any ipsilateral lung failure was considered a local radiotherapy (RT) treatment planning. As discussed
failure. However, when using a tighter definition of previously, appropriate treatment and prognosis
local failure (ipsilateral hilum/mediastinum or bron- depend on accurate staging; even now, clinical stag-
chial stump—areas typically covered by a conformal ing can be difficult and unreliable. High quality
radiotherapy portal) the local failure rate was only 16%. images from CT to PET scans also help in planning
Sawyer et al. (1997) conducted a retrospective areas for radiation treatment. There have also been
review of 224 patients who underwent complete improvements in radiation source intensity and
resection of N2 NSCLC at the Mayo Clinic between application techniques that have reduced morbidity
Postoperative Radiotherapy for Non-Small Cell Carcinoma 365

and mortality by treating smaller and often irregular stage I NSCLC. Most randomized trials in node
volumes with smaller margins (Le Pechoux 2011). positive NSCLC suggest that PORT improves loco-
In 1980, Van Houtte et al. completed a randomized regional control though effect on survival has been
trial with 175 patients with pathological stage I unclear (Weisenburger 1994; Stephens et al. 1996;
NSCLC and no node involvement who received Dautzenberg et al. 1999; Feng et al. 2000; Mayer
complete resections. PORT was found to have a et al. 1997). The Dautzenberg paper (which used
detrimental effect on overall survival, with 5-year suboptimal radiotherapy techniques and doses) was
overall survival rates of 24% with PORT, compared strongly ‘‘negative’’ against PORT, but most other
to 43% in the control group. Van Houtte et al. rea- studies are underpowered to make any conclusions
sonably concluded that thoracic radiation therapy about PORT in node-positive NSCLC.
should not be performed on N0 NSCLC following Given the shortcomings of the randomized trials
surgical resection (Van Houtte et al. 1980). This was (Table 1), it is appropriate to review lower level of
confirmed in the PORT meta-analysis (PORT meta- evidence papers (retrospective reports). There are
analysis trialists Group 1998). many retrospective studies showing that PORT
Trodella et al’s controlled randomized study pub- improves survival for node-positive NSCLC. For
lished in 2002 was one of the few studies to suggest a example, Dai et al. (2011) performed a retrospective
benefit of PORT in stage I NSCLC. In contrast to the study of 221 cases in a Beijing hospital and found that
above listed trials, treatments were planned with CT, and PORT can significantly improve the survival as well as
radiotherapy was administered with linear accelerator local and distant recurrences of patients with resected
and 3D beam angles. The patients with pathological pathological stage IIIA-N2 NSCLC. Five-year overall
stage I NSCLC all had complete resection, treated survival of the 96 patients (43.4%) who received PORT
between 1989 and 1997. The study concluded that was 36.6%, compared to 30.6% of patients who did not
PORT resulted in lower local recurrence rate, without receive PORT. PORT and non-PORT groups had sta-
acute toxicity or negative impact on overall or disease- tistically significantly different overall survival and
free survival, and with a positive trend (not statistically disease-free survival. PORT was found to be a signifi-
significant) in survival. The authors endorsed further cant positive prognostic factor and an independent
investigation of PORT, particularly with advances in prognostic factor for overall survival in univariate and
radiotherapy technique (Trodella et al. 2002). multivariate analyses. Furthermore, locoregional
The PORT Meta-analysis Trialists Group’s upda- recurrence-free survival (LRFS) and distant metasta-
ted meta-analysis included results of 10 randomized sis-free survival (DMFS) rates were significantly
trials, including that of Trodella et al, with a total of higher in the PORT than in the non-PORT groups, with
2232 cases of NSCLC. The meta-analysis concluded 5-year LRFS/DMFS rates of 63.9/43.8% and 46.7/
that PORT has a detrimental effect on survival of 23.6%, respectively. Of the 221 cases in this study, 161
Stage I (and II) NSCLC patients (Burdett et al. 2005). (72.9%) also received adjuvant chemotherapy while 60
For now, the general body of evidence seems to (27.1%) did not; PORT improved survival significantly
suggest that PORT may be more detrimental than in both these groups considered separately. These
beneficial in stage I NSCLC (Table 1). Indeed, some results appear to confirm the data from other recent
clinicians may argue that the risk for recurrence of retrospective reports (Zou et al. 2010; Moretti et al.
stage I disease is not high enough to warrant PORT 2009; Scotti et al. 2010).
(Le Pechoux 2011). Re-evaluation may be necessary Douillard et al. published a secondary analysis of
as RT techniques improve over time. the large ‘‘ANITA’’ (adjuvant chemo) trial (Douillard
et al. 2008). They performed a secondary analysis of
the large adjuvant navelbine international trialist
4 Results of PORT in Stage II and III association (‘‘ANITA’’) randomized trial. Without
(Node-Positive) NSCLC considering pathologic N stages separately, PORT
groups had shorter 5-year overall survival (33% in
Notwithstanding the results of the PORT meta-anal- observation and 44.6% in adjuvant chemotherapy
ysis Trialists Group results, the situation for node- group) than the non-PORT groups (43% in observa-
positive (stage II/III) NSCLC is less clear than for tion and 51% in adjuvant chemotherapy group).
366 E. Kim and M. Machtay

Table 1 Results of selected randomized trials of postoperative radiotherapy (PORT) for NSCLC
Study # patients Stage XRT dose Survival with Survival without LRF with LRF without
(Gy) XRT (%) XRT (%) XRT (%) XRT (%)
Van Houtte et al. (1980) 202 I–III 60 24a 43a 2 11
Weisenburger (1994) 230 II, III 50 40 40 3a 21a
Feng et al. (2000) 317 II, III 60 43 41 13a 33a
Lafitte et al. (1996) 163 I 45–60 35 52 15 17
a
Stephens et al. (1996) 308 II, III 40 25 25 18 29a
Mayer et al. (1997) 155 I–III 50–56 30 20 6 24
a a
Dautzenberg et al. (1999) 720 I–III 60 30 43 28 34
b b
Debevec et al. (1996) 74 III 30 32 20
Trodella et al. (2002) 104 I 50 67a 58a 2a 22a
a
Statistically significant difference (P \ 0.05)
b
Data not available

However, quite different results were found when to the negative results of the 1998 meta-analysis (1998);
pathologic N1 and N2 stages were considered sepa- however, the rationale for PORT remains the same as
rately. There was an insufficient quantity of cases to for radiotherapy for other malignancies. First, patients
make any conclusions about pathologic N0 cases. at high risk for locoregional recurrence can be identi-
Considering only pathologic N1 cases, PORT was fied; second, properly and appropriately used radio-
found to have better survival than observation alone, therapy can improve locoregional control; third, the
but PORT and adjuvant chemotherapy was found to benefits of locoregional control outweigh the toxicities
have poorer survival than adjuvant chemotherapy of radiation so that overall, it is clinically beneficial to
alone. Considering only pathologic N2 cases, for both the patient to undergo radiotherapy (Machtay 2008).
observation and adjuvant chemotherapy groups, Radiotherapy in the context of PORT for NSCLC
patients who received PORT had better survival than fulfills the first two of these reasons. However, the third
those in the same group who did not receive PORT. hypothesis requires further investigation. Current
The Douillard and other retrospective papers can randomized trials that may provide more guidance are
be understandably criticized as falling short of the still ongoing (Dai et al. 2011; Le Pechoux 2011).
high level of evidence demanded by modern evalua- The following are patient selection criteria based on
tors of medical therapeutics. A small randomized trial information presently available. Patients with N2 dis-
in the United States in which patients were assigned ease should receive adjuvant chemotherapy, based on
to adjuvant chemotherapy with or without PORT level 1 evidence of improved survival. PORT should be
closed early due to suboptimal accrual. Prospective strongly considered for these patients as well (Machtay
randomized multi-center clinical trials are currently 2008; Decker 2008). Physicians may want to consider
ongoing in France and China (Le Pechoux 2011; the extent of mediastinal adenopathy in the decision
Dai et al. 2011). A proposal for attempting another process. A retrospective study by Matsugama et al.
similar randomized trial in the U.S. was considered (2008) suggested that PORT was more effective for
infeasible. multiple station metastases than single station metas-
tasis. Thus, though prospective randomized trials are
warranted, these results suggest that PORT can reduce
5 Indications for PORT local recurrence and improve overall survival, at least
for patients with multiple station N2.
There is currently no consensus for which patients Patients should have good performance status,
should receive PORT (Machtay 2008). The use of good cardiopulmonary reserve and pre-PORT organ
PORT decreased during the 2000s, probably in reaction function testing (Machtay 2008). Patients should also
Postoperative Radiotherapy for Non-Small Cell Carcinoma 367

be willing and able to comply with careful intra- and information from randomized trials, Kelsey, Marks,
post-treatment medical care and visits (Machtay and Wilson noted that most non-cancer deaths occur
2008). Ideally, PORT should be applied using the best close to the first 2 years after completion of therapy,
technology available, such as 3D conformal (CRT), and are related to cardiopulmonary disease. Kelsey,
intensity modulated (IMRT), and/or proton-beam Marks, and Wilson conjectured that acute toxicities
radiotherapy, limiting radiation dose to adjacent vital generally assumed to be transient, such as esophagitis,
organs and using a target dose B54 Gy (Machtay et al. may actually predispose the patients to lethal respi-
2001; Machtay 2008). ratory and cardiac complications (Kelsey et al. 2008).
Several studies have provided evidence that PORT
toxicity has decreased with improvements in planning
6 Toxicity and Mortality Risks and treatment technology. Lally et al. (2007) conducted
of PORT a large retrospective study of 6,148 patients diagnosed
with ipsilateral lymph node positive NSCLC between
For at least 30 years, PORT has been suggested to 1983 and 1993 who received surgery (pneumonectomy
improve local control for patients with resected or lobectomy) using Surveillance, Epidemiology, and
NSCLC (Van Houtte et al. 1980; Weisenburger 1994; End Results (SEER) data. They investigated the impact
Stephens et al. 1996; Mayer et al. 1997; Keller et al. of PORT on cardiac mortality, the main cause of death
2000; Trodella et al. 2002), but a major concern is from intercurrent disease due to irradiation of heart
that toxicity outweighs this survival benefit. Most of tissue. PORT was found to increase the risk of cardiac
the time, the toxicities of PORT are transient and mortality with statistical significance for patients
modest in intensity (radiation dermatitis, esophagitis). diagnosed between 1983 and 1988, but not for patients
However, more serious adverse events can occur, and diagnosed between 1988 and 1993.
cause of death can be difficult to determine, especially Machtay et al. (2001) reviewed 202 patients
when patients have pre-existing medical conditions diagnosed with pathologic stage II or III NSCLC
and other comorbidities of smoking (Kelsey et al. between 1982 and 1998 and found an actuarial rate of
2008). Dautzenberg et al. (1999) showed significantly death from intercurrent disease (DID) to be 13.5%,
increased risk of death from non-cancer causes in statistically insignificantly greater than the estimated
patients randomized to receive PORT. 10% for a matched population. Further investigation
In the well-known Lung Cancer Study Group revealed that DID was associated with higher total
(LCSG) study of 230 patients randomized to PORT or radiation dose; the risk of DID was 2% for patients
observation, Weisenburger (1994) also published the with dose \54 Gy, compared to 16% for patients who
details of toxicities. In the PORT group, they noted received higher doses. This reinforced the hypothesis
that 24% of patients had serious esophagitis, 20% had that a higher incidence of DID was significantly
gastrointestinal symptoms, 11% had dermatologic, associated with higher daily doses of radiation
and 10% had neurologic toxic reactions. PORT and (Le Pechoux 2011). It should be noted that the
control groups had statistically non-different pul- Dautzenberg trial used a high total dose (60 Gy) and
monary toxic reactions. Two patients in the PORT fraction size (2.5 Gy) of radiation.
group, compared to one patient in the control group, In a similar study, Wakelee et al. (2005) analyzed the
experienced adverse events that were so severe as to risk of DID in stage II and IIIA NSCLC patients
be life threatening. It must be noted that these patients following surgical resection and either PORT or che-
were randomized between 1978 and 1985, so it is moradiotherapy. In their randomized trial of 488
possible that they overestimate the current toxicities patients, 242 patients received PORT (50.4 Gy in 28
associated with modern treatment planned PORT daily fractions) and 246 patients received PORT with
(Saynak et al. 2010). cisplatin and etoposide administered concurrently. They
The greatest concern about PORT is that it may concluded that the actuarial overall 4-year DID rate was
cause death due to pulmonary and/or cardiac deteri- not significantly different between the two groups.
oration even in the absence of a recognized treat- Masson-Cote et al. (2011) conducted a retrospec-
ment-related toxicity event (Machtay et al. 2001; tive chart review of 153 lung cancer patients who
Dautzenberg et al. 1995, 1999). Based on published received PORT between 1995 and 2007, examining
368 E. Kim and M. Machtay

the differences in locoregional control and survival to mathematically relate survival with tumor stage
between patients who were treated with 2-dimen- and field size based on the available clinical data.
sional (2D) compared with 3D PORT. Kaplan–Meier Exploring the balance of benefits of cancer-specific
analysis showed that the 3D technique had far survival afforded by PORT and risks of radiation-
superior results than 2D technique, with a 5-year induced mortality, they found that radiation-induced
locoregional control rate of 81% compared to 56%. mortality seems proportional to the cube of the field
Toxicity was similar and modest in both groups. size. The changes in survival predicted by this
A recent retrospective study by Dai et al. (2011) of formula corresponded to survivals reported in the
patients with pathologic stage IIIA-N2 NSCLC cate- available literature.
gorized the causes of death. A total of 221 patients A full review of the technical details for radiation
were diagnosed between 2003 and 2005; 96 (43.4%) treatment planning for PORT is beyond the scope of
patients received PORT, and 140 had died by the time this chapter, but the important factors in planning
of the last follow-up in 2009. One hundred twenty- PORT Radiotherapy are: (1) Adequate coverage of
three (87.9%) of the 140 deaths were cancer related, the area(s) at high risk of local recurrence (bronchial
including 53 who had received PORT and 70 who had stump, hilum, partial mediastinum and (2) protection
not. Seventeen (12.1%) of the 140 deaths were of critical normal lung and heart tissue from excessive
categorized as non-cancer-related and were evenly irradiation. 3D conformal RT with detailed attention
distributed in the PORT and non- PORT groups. to dose-volume histograms (DVH) is critical to the
Technologic advances have improved staging, safe planning and delivery of PORT. The target vol-
planning, and control of radiation administration, ume should generally not be irradiated [54 Gy and
sparing more normal tissue, so that modern PORT for the lung V20 (volume % of lung receiving C20 Gy)
NSCLC does not significantly increase the risk of must be minimized, preferably B25%.
intercurrent deaths (Machtay et al. 2001). This redu-
ces the risks of PORT in NSCLC treatment, reducing
one of the primary reasons against PORT. Further 8 PORT and Adjuvant Chemotherapy
investigation will be required to find the optimal
conditions and treatments for PORT. As noted above, adjuvant chemotherapy has become
the standard of care for stages II–IIIA NSCLC. This is
largely based on the results of three randomized trials
7 Techniques for PORT of cisplatin-based adjuvant chemotherapy regimens as
given by Arriagada et al. (2004), Douillard et al.
As noted above, PORT has a strong potential to (2008), and Winton et al. (2005) that showed that
increase local control (and thereby survival), and also cisplatin-based adjuvant chemotherapy improved
to cause serious toxicities, even cardiopulmonary survival among patients with completely resected
death. It is thus critical that meticulous radiation NSCLC. The impact of adjuvant chemotherapy on
therapy treatment planning be employed. locoregional control is less well-documented in the
One of the criticisms of the older series of PORT is current literature (Saynak et al. 2010).
that they used Cobalt-60 radiotherapy beams rather The balance of risks and benefits of PORT fol-
than linear accelerator-based RT. Among other limi- lowing surgical resection remains more unclear, and
tations, Co-60 beams have a wide ‘‘penumbra,’’ thus the sequencing of adjuvant chemo and PORT is
meaning that radiotherapy scatter dose outside the unclear. A randomized trial of 488 clinical stage II or
radiotherapy portal is substantially greater than linear IIIA NSCLC patients by Keller et al. (2000) con-
accelerator-based RT. Phlips et al. (1993) showed that cluded that concurrent cisplatin-based chemotherapy
patients treated with PORT using linear accelerator along with PORT did not improve either locoregional
had a significantly better outcome than patients trea- control or survival when compared to PORT alone
ted with Co-60. after surgery. The Radiation Therapy Oncology
Volume irradiated likely depends not only on the Group (Bradley et al. 2005) reported that their study
prescribed radiotherapy dose, but also on the volume of 88 patients with surgical resection for pathologic
of tissue irradiated. Miles et al. (2007) created models stage II or IIIA NSCLC patients who received
Postoperative Radiotherapy for Non-Small Cell Carcinoma 369

postoperative concurrent paclitaxel/carboplatin and lung cancer: promising long-term results of the Radiation
PORT found the combination to be safe and that it Therapy Oncology Group–RTOG 9705. J Clin Oncol 23(15):
3480–3487. PMID: 15908657
seemed to have an improved overall and progression- Burdett S, Stewart L, PORT Meta-analysis Group (2005)
free survival compared to previously reported trials. Postoperative radiotherapy in non-small-cell lung cancer:
This suggests that further study is warranted. update of an individual patient data meta-analysis. Lung
However, at this time, concurrent chemo-RT in the Cancer 47(1):81–83. PMID: 15603857
Dai H, Hui Z, Ji W (2011) Postoperative radiotherapy for
adjuvant setting is thus not routinely recommended. resected pathological stage IIIA-N2 non-small cell lung
Most centers prefer a sequence in which adjuvant cancer: a retrospective study of 221 cases from a single
chemo is given first, followed by PORT. institution. Oncologist 16(5):641–650. Epub 2011 Apr 11,
PMID: 21482587
Dautzenberg B, Chastang C, Arriagada R et al (1995) Adjuvant
radiotherapy versus combined sequential chemotherapy fol-
9 Conclusions and Future Directions lowed by radiotherapy in the treatment of resected nonsmall
cell lung carcinoma. A randomized trial of 267 patients.
Local–regional failure after surgery remains an GETCB (Groupe d’Etude et de Traitement des Cancers
Bronchiques). Cancer 76(5):779–786. PMID: 8625180
important problem in the management of NSCLC. Dautzenberg B, Arriagada R, Chammard AB et al (1999) A
The most significant factor predictive for local– controlled study of postoperative radiotherapy for patients
regional failure is node positivity, particularly N2 with completely resected nonsmall cell lung carcinoma.
(mediastinal) adenopathy. It is generally agreed that Groupe d’Etude et de Traitement des Cancers Bronchiques.
Cancer 86(2):265–273. PMID: 10421262
PORT reduces the risk of local–regional failure sub- Debevec M, Bitenc M, Vidmar S et al (1996) Postoperative
stantially; however the effect of PORT on overall radiotherapy for radically resected N2 non-small-cell lung
survival is unclear. It is possible that the beneficial cancer: randomized clinical study 1988–1992. Lung Cancer
effects of PORT are outweighed in some patient 14(1):99–107. PMID: 8696724
Decker RH, Wilson LD (2008) Postoperative radiation therapy
groups by morbidity and mortality from PORT. for non-small cell lung cancer. Semin Thorac Cardiovasc
Although there are some international randomized Surg 20(3):184–187. PMID: 19038726
trials re-investigating the use of PORT, it is unlikely Douillard JY, Rosell R, De Lena M et al (2008) Impact of
that definitive conclusions will be reached very soon. postoperative radiation therapy on survival in patients with
complete resection and stage I, II, or IIIA non-small-cell lung
Thus, current recommendations are that most patients cancer treated with adjuvant chemotherapy: the adjuvant
with Stage I and II disease not receive PORT (unless Navelbine International Trialist Association (ANITA)
there is evidence of residual cancer). Patients with Randomized Trial. Int J Radiat Oncol Biol Phys 72(3):695–
Stage III (N2) disease should be offered PORT, rec- 701. Epub 2008 Apr 24.18439766
Feng QF, Wang M, Wang LJ et al (2000) A study of
ognizing that adjuvant chemotherapy should be the postoperative radiotherapy in patients with non-small-cell
higher priority for these patients because of the lat- lung cancer: a randomized trial. Int J Radiat Oncol Biol
ter’s greater effect on overall survival. There is still Phys. 47(4):925–929. PMID: 10863061
much to learn about patterns of failure and clinical Keller SM, Adak S, Wagner H et al (2000) A randomized trial of
postoperative adjuvant therapy in patients with completely
and biologic factors that predict for local–regional resected stage II or IIIA non-small-cell lung cancer. Eastern
failure after surgery. Future studies to assess this are Cooperative Oncology Group. N Engl J Med 343(17):
needed in order to refine the precise indications for 1217–1222. PMID: 11071672
PORT and how to integrate PORT with adjuvant Kelsey CR, Light KL, Marks LB (2006) Patterns of failure after
resection of non-small-cell lung cancer: implications for
chemotherapy. postoperative radiation therapy volumes. Int J Radiat Oncol
Biol Phys 65(4):1097–1105. Epub 2006 May 6, PMID:
16682136
References Kelsey CR, Marks LB, Wilson LD (2008) Postoperative
radiation therapy for lung cancer: where do we stand?
Oncology (Williston Park) 22(3):301–310; discussion 310,
Arriagada R, Bergman B, Dunant A et al (2004) Cisplatin- 314–315, 319. PMID: 18494356
based adjuvant chemotherapy in patients with completely Lafitte JJ, Ribet ME, Prevost BM et al (1996) Postresection
resected non-small-cell lung cancer. N Engl J Med 350(4): irradiation for T2N0M0 non-small cell carcinoma: a
351–360. PMID: 14736927 prospective randomized trial. Ann Thorac Surg 62(3):
Bradley JD, Paulus R, Graham MV et al (2005) Phase II trial of 830–834. PMID: 8784014
postoperative adjuvant paclitaxel/carboplatin and thoracic Lally BE, Detterbeck FC, Geiger AM et al (2007) The risk of
radiotherapy in resected stage II and IIIA non-small-cell death from heart disease in patients with nonsmall cell lung
370 E. Kim and M. Machtay

cancer who receive postoperative radiotherapy: analysis of Saynak M, Higginson DS, Morris DE et al (2010) Current status
the surveillance, epidemiology, and end results database. of postoperative radiation for non-small-cell lung cancer.
Cancer 110(4):911–917. PMID: 17620279 Semin Radiat Oncol 20(3):192–200. PMID: 20685582
Le Pechoux C (2011) Role of postoperative radiotherapy in Scotti V, Meattini I, Saieva C et al (2010) Post-operative
resected non-small cell lung cancer: a reassessment based radiotherapy in N2 non-small cell lung cancer: a retrospec-
on new data. Oncologist 16(5):672–681. Epub 2011 Mar 4, tive analysis of 175 patients. Radiother Oncol 96(1):84–88.
PMID: 21378080 Epub 2010 Jun 11, PMID: 20541823
Machtay M (2008) PORTable indications in non-small-cell Stephens RJ, Girling DJ, Bleehen NM (1996) The role of
lung carcinoma. Oncology 22:301–310 postoperativef radiotherapy in non-smalol cell lung cancer:
Machtay M, Lee JH, Shrager JB et al (2001) Risk of death from a multicentre randomised trial in patients with pathologi-
intercurrent disease is not excessively increased by modern cally staged T1-2N1-2M0 disease. Medical research council
postoperative radiotherapy for high-risk resected non-small- lung cancer working party. Br J Cancer 74(4):632–639.
cell lung carcinoma. J Clin Oncol 19(19):3912–3917. PMID: 8761382
PMID: 11579111 Strauss GM, Herndon JE II, Maddaus MA et al (2008)
Masson-Cote L, Couture C, Fortin A et al (2011) Postoperative Adjuvant paclitaxel plus carboplatin compared with obser-
radiotherapy for lung cancer: improvement in locoregional vation in stage IB non-small-cell lung cancer: CALGB 9633
control using three-dimensional compared with two- with the Cancer and Leukemia Group B, Radiation Therapy
dimensional technique. Int J Radiat Oncol Biol Phys 80(3): Oncology Group, and North Central Cancer Treatment
686–691 Group Study Groups. J Clin Oncol 26(31):5043–5051. Epub
Matsugama H, Nakahara R, Ishikawa Y et al (2008) Postop- 2008 Sept 22, PMID: 18809614
erative radiotherapy for patients with completely resected Taylor NA, Liao ZX, Stevens C et al (2003) Postoperative
pathological stage IIIA-N2 non-small cell lung cancer: radiotherapy increases locoregional control of patients with
focusing on an effect of the number of mediastinal lymph stage IIIA non-small-cell lung cancer treated with induction
node stations involved. Interact Cardiovasc Thorac Surg chemotherapy followed by surgery. Int J Radiat Oncol Biol
7(4):573–577. Epub 2008 Apr 15, PMID: 18413349 Phys 56(3):616–625. PMID: 12788166
Mayer R, Smolle-Juettner FM, Szolar D et al (1997) Postop- Trodella L, Granone P, Valente S et al (2002) Adjuvant
erative radiotherapy in radically resected non-small cell radiotherapy in non-small cell lung cancer with pathological
lung cancer. Chest 112(4):954–959. PMID: 9377958 stage I: definitive results of a phase III randomized trial.
Miles EF, Kelsey CR, Kirkpatrick JP et al (2007) Estimating Radiother Oncol 62(1):11–19. PMID: 11830308
the magnitude and field-size dependence of radiotherapy- Van Houtte P, Rocmans P, Smets P et al (1980) Postoperative
induced mortality and tumor control after postoperative radiation therapy in lung caner: a controlled trial after
radiotherapy for non-small-cell lung cancer: calculations resection of curative design. Int J Radiat Oncol Biol Phys
from clinical trials. Int J Radiat Oncol Biol Phys 6(8):983–986. PMID: 6998936
68(4):1047–1052. Epub 2007 May 15, PMID: 7507176 Varlotto JM, Medford-Davis LN, Recht A (2010) Varying
Moretti L, Yu DS, Chen H et al (2009) Prognostic factors for recurrence rates and risk factors associated with different
resected non-small cell lung cancer with pN2 status: definitions of local recurrence in patients with surgically
implications for use of postoperative radiotherapy. resected, stage I nonsmall cell lung cancer. Cancer
Oncologist 14(11):1106–1115. Epub 2009 Nov 6, PMID: 116(10):2390–2400. PMID: 20225332
19897534 Wakelle HA, Stephenson P, Keller SM et al (2005) Post-
NSCLC Meta-analyses Collaborative Group, Arriagada R, operative radiotherapy (PORT) or chemoradiotherapy
Auperin A et al (2010) Adjuvant chemotherapy, with or (CPORT) following resection of stages II and IIIA non-
without postoperative radiotherapy, in operable non-small- small cell lung cancer (NSCLC) does not increase the
cell lung cancer: two meta-analyses of individual patient expected risk of death from intercurrent disease (DID) in
data. Lancet 375(9722):1267–1277. Epub 2010 Mar 24, Eastern Cooperative Oncology Group (ECOG) trial E3590.
PMID: 20338627 Lung Cancer 48(3):389–397. Epub 2005 Jan 5, PMID:
Phlips P, Rocmans P, Vanderhoeft P et al (1993) Postoperative 15893008
radiotherapy after pneumonectomy: impact of modern Weisenburger TH (1994) Effects of postoperative mediastinal
treatment facilities. Int J Radiat Oncol Biol Phys 27(3): radiation on completely resected stage II and stage III
525–529. PMID: 8226144 epidermoid cancer of the lung. LCSG 773. Chest 106(6
PORT Meta-analysis Trialists Group (1998) Postoperative Suppl):297S–301S. PMID: 7988248
radiotherapy in non-small-cell lung cancer: systematic Winton T, Livingston R, Johnston D et al (2005) Vinorelbine
review and meta-analysis of individual patient data from plus cisplatin vs. observation in resected non-small-cell
nine randomised controlled trials. Lancet. 352(9124): lung cancer. N Engl J Med 352(25):2589–2597. PMID:
257–263. PMID: 9690404 15972865
Sawyer TE, Bonner JA, Gould PM et al (1997) The impact of Zou B, Xu Y, Li T et al (2010) A multicenter retrospective
surgical adjuvant thoracic radiation therapy for patients with analysis of survival outcome following postoperative che-
nonsmall cell lung carcinoma with ipsilateral mediastinal moradiotherapy in non-small-cell lung cancer patients with
lymph node involvement. Cancer 80(8):1399–1408. PMID: N2 nodal disease. Int J Radiat Oncol Biol Phys 77(2):
9338463 321–328. Epub 2009 Sept 21, PMID: 19775829
PDT-Lung
Ron R. Allison

Contents Abstract
The role of Photodynamic Therapy (PDT) in the
1 Introduction............................................................ 371 current multi-disciplinary approach to thoracic
2 Historical Perspective............................................ 372 oncology is analyzed and reviewed. The simplicity
of drug, light and reaction resulting in excellent
3 Fundamentals of PDT ........................................... 372
3.1 Photosensitizer ......................................................... 372 clinical response has brought PDT to a worldwide
audience. This chapter reviews the scientific and
4 Illumination ............................................................ 373
clinical rationale for thoracic PDT including
5 Photodynamic Reaction ........................................ 373 photosensitizers, light sources, photodynamic reac-
6 Dosimetry................................................................ 374 tion, dosimetry and fluorescence. The actual treat-
ment procedure, clinical outcomes from the peer
7 Fluorescence and Photo Diagnosis ...................... 374
reviewed literature, limitations and morbidities
8 Rationale for Pulmonary PDT............................. 374 associated with this light-based therapy are also
9 PDT Procedure ...................................................... 375 detailed.
10 PDT-Morbidity....................................................... 376
11 Clinical Outcomes.................................................. 376
11.1 Obstructing Endobrochial Lesions.......................... 376
11.2 Early Lung Cancer .................................................. 377 1 Introduction
11.3 Multiple Primary Lung Cancer ............................... 377
11.4 Down Staging .......................................................... 377 Photodynamic Therapy (PDT) has a long clinical
11.5 Peripheral Lesions ................................................... 378
track record of success in the treatment of pul-
11.6 Pleural Tumors ........................................................ 378
11.7 PDT in Combination with Radiation...................... 378 monary malignancy (Allison et al. 2004a, b).
The simplicity of this intervention in combination
12 Conclusion .............................................................. 378
with high-response rates has brought PDT to a
References.......................................................................... 378 worldwide audience (Dougherty and Marcus 1992).
Further PDT can serve as a model therapy for
tumor ablation in which functional preservation is
often achieved without undue acute or chronic
toxicity. This chapter will review the fundamental
scientific basis of PDT, treatment procedures and
precautions, clinical outcomes and indications and,
ultimately, the place of PDT in the modern multi-
R. R. Allison (&)
Twenty-first Century Oncology, 801 WH Smith Blvd,
disciplinary decision-making process and therapy
Greenville, NC 27834, USA of lung cancer.
e-mail: rallison@rtsx.com

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 371
DOI: 10.1007/174_2011_220, Ó Springer-Verlag Berlin Heidelberg 2011
372 R. R. Allison

PDR a rapid vascular shutdown is often noted as


2 Historical Perspective necrosis and/or apoptosis of the target tissue. The
clinical result is lesion ablation with excellent cos-
PDT was discovered serendipitously by Oscar Raab, a metic and functional treatment outcome. To better
medical student (Raab 1900). Raab was examining understand the PDT process we will examine each
the fluorescent characteristics of dyes on infusoria. component, as described below.
Raab noted and examined the unexpected deaths of
these microorganisms when the dye was illuminated
intensely. In combination with his colleagues and 3.1 Photosensitizer
professor, notably Von Tappenier, Jesionek and
Joldlbauer, the oxygen-dependent nature of this The PS transfers light energy to create what is termed
Photodynamic Reaction (PDR) was elucidated and a type II photochemical reaction which results in
this new form of light-based treatment rapidly rapid generation of highly destructive singlet oxygen
developed (Von Tappeiner and Jesionek 1903). By radicals (Allison and Sibaba 2010). As photosynthe-
1907 a textbook of PDT was available and PDT sis, another form of light energy transfer, is critical to
offered excellent ablation of various forms of cuta- life on earth, it should not be surprising that numerous
neous malignancy (Von Tappeiner and Jesionek natural and synthetic substances are able to transfer
1907). Still, PDT did not firmly establish itself and light energy. But to be considered a PS the light
was lost to medicine. The use of dyes and later por- energy transfer must result in a type II reaction. While
phyrins, for fluorescent detection, as potential radia- thousands of potential PS agents exist only a handful
tion sensitizers and, ultimately, for PDT was again has been brought through clinical trials and even
seen intermittently in the scientific literature during fewer are commercially available for therapy.
the 1950s and early 1960s (Allison et al. 2004a, b; A clinically successful PS will have some or all of
Daniell and Hill 1991). However, it was not until the the following characteristics: (Allison and Sibaba
tireless efforts of Dougherty in the 1970s that PDT 2010) ability to concentrate in target tissue, clearance
achieved a worldwide audience (Dougherty 1996). In from non-target normal tissue, reliable reactivity only
contrast to prior attempts, Dougherty spearheaded the upon illumination, relatively rapid clearance from the
development of a clinically versatile and commer- subject, pain-free therapy, ease of drug delivery
cially successful photosensitizing drug, light sources (topical, oral, IV), controllable PDR, synthetic ease
and clinical trials showing efficacy in a wide variety and purity, reproducible formulation and non-toxicity
of malignancies particularly pulmonary lesions. Dur- until activated. It must also have regulatory approval
ing the early 1980s PDT achieved worldwide regu- and commercial availability. Many outstanding PS
latory approval as an intervention for select thoracic have been discovered but due to lack of commercial
tumors. Since those early approvals, the PDT treat- production they are not available to help patients.
ment process has been refined and enhanced allowing Not surprisingly the porphyrins, which form the
for continued success today. back bone of chlorophyll and hemoglobin, two highly
successful structures for transfer of oxygen, have
served as the basis for clinically successful PS
3 Fundamentals of PDT development. In particular, two PS agents have found
success in pulmonary PDT.
Arbitrarily we will divide PDT into three compo- PhotofrinÒ—This proprietary mixture of porphyrin
nents: Photosensitizing agent (PS), light source monomers, dimers and oligomers was the product of
and photodynamic reaction (PDR) (Allison et al. Dr. Dougherty’s pioneering work in PDT and has
2004a, b). In reality creating a successful PDR remained the most commonly utilized PS agent for
requires appropriate localization and accumulation of more than 30 years. PhotofrinÒ is commercially
PS in the target tissue followed by illumination of this available from Axcan Pharma but is also manufac-
PS with a specific intensity and wavelength of light. tured by numerous ‘‘unofficial’’ manufacturers whose
All of this must coalesce in time and space to generate drug appears similar to PhotofrinÒ in both name and
the oxygen-dependent PDR. Upon creation of the content. This PS allows for treatment time of
PDT-Lung 373

10–20 min per lesion which is satisfactory during


bronchoscopy. Therapy is painless and with photo-
frinÒ highly reliable. It is usually dosed at 2 mg/kg
and around 48 h post intravenous infusion a differ-
ential concentration exists between pulmonary tumors
and surrounding normal tissue. This may allow for
permanent lesion ablation without permanent normal
tissue damage. The major drawback is that enough
PhotofrinÒ is retained in normal tissue, including the
skin, for 6 weeks so that unintentional sunlight
exposure during this time frame will create a poten-
tially significant burn in the exposed skin.
MACEÒ—a chlorine-based PS. Mono-l-aspartyl
Chlorine e6 has recently achieved regulatory status in
Japan for pulmonary PDT. This synthetically pure PS
Fig. 1 The Photodynamic Reaction. The active photosensi-
has a more rapid clearance from normal tissue and
tizer (PS) may follow several pathways including one that
skin allowing for a 2–3 week period of photosensi- generates toxic singlet oxygen (PDR)
tivity. A differential between normal tissue and tumor
is noted at 4 h post intravenous infusion allowing for
same day infusion and PDT as compared to the two- A flashlight-like forward projecting micro lens fiber
day waiting period for photofrin. The drug is intra- optic is commercially available as is a diffusion fiber
venously infused at 40 mg/m2. which illuminates in all directions. Recently Light
Emitting Diodes (LED) tuned to the PS wavelength
have become available and are much less expensive
4 Illumination both in purchase outlay and use than lasers. On the
forefront of illumination are highly portable battery
Each PS has both a specific activation wavelength and operated light sources allowing for prolonged or
also intensity of light required for activation. As sun- repeated (metronomic) illumination protocols.
light contains multiple wavelengths and is also an
intense source of illumination energy it can activate
any PS. Since all current PS accumulate to a certain 5 Photodynamic Reaction
degree in the skin avoiding sunlight exposure is a
critical aspect in minimizing morbidity. Clinically The successful culmination of PS activation by the
useful wavelengths for PS activation are in the red light appropriate intensity and wavelength of illumination
range (600 nm and above). Red light penetrates tissue is the PDR (Allison and Sibaba 2010). This is illus-
to about 1 cm which is more than satisfactory for trated in Fig. 1. As mentioned this is the generation of
treatment of in situ and non-bulky invasive pulmonary toxic singlet oxygen as a product of the type II (PDR)
lesions. PhotofrinÒ activates at 630 nm and MACEÒ at reaction. In this process the inert PS is converted to
664 nm. Wavelengths above 800 nm are absorbed the active triplet energy state via proton transfer from
preferentially by water so the illumination window by light. The PS will transfer the energy which cleaves
light for successful PDT is rather limited. More bulky an oxygen molecule ultimately generating the singlet
lesions can be treated through repeated PDT sessions oxygen. The active PS can also generate a toxic type I
or via interstitial placement of the light source within reaction via generation of hydroxyl radicals cleaved
the tumor. Currently portable diode lasers are able to from water. A direct cytotoxic, non-oxygen mediated
generate enough power to activate the PS (Mang et al. type III reaction may also occur. An additional
1987). A laser tuned to the specific activation wave- pathway is that the active PS may lose energy via
length of PhotofrinÒ or MACEÒ is commercially release of light, termed fluorescence. This visible
available. These lasers are coupled to fiber optics that release of light can be of great clinical utility as will
are brought to the region requiring illumination. be described later.
374 R. R. Allison

Clinically with the intense illumination currently based on the patient’s metabolic and treatment char-
employed the PDR presents with vascular shutdown acteristics. This is an area of active research.
and a hypoxic appearing lesion. The type II reaction
generally results in cellular necrosis via destruction of
cell and vascular membranes which is where the PS 7 Fluorescence and Photo Diagnosis
accumulates. As PS does not accumulate in DNA,
PDT is considered a non-mutagenic therapy. The As mentioned one pathway for PS activation leads to
release of cellular debris initiates a cytokine cascade the release of visible or detectable light, termed
which may potentiate an immune-type response. In fluorescence (Allison and Sibaba 2010). The fluores-
contrast, an apoptotic death appears favored from a cence phenomena can be exploited to assist in
prolonged or metronomic illumination. Manipulating determining the extent of disease which may be
the PDR is an area of active research. unclear to the naked eye in visible light. Under fluo-
rescence lesions become very obvious compared to
nearby non-fluorescing normal tissues. This fluores-
6 Dosimetry cence phenomenon is sometimes called photo diag-
nosis. Change in the fluorescence before and after
Light dosimetry is far more complicated than, for PDT may also be a means of dosimetry. Full loss of
example, dosimetry in radiation therapy (Sibata et al. fluorescence can indicate tumor death. Thus treatment
2000). The interaction of light with matter remains to could be individualized based on loss of fluorescence
be better defined. However, clinicians use PS drug rather than by using the current methods of dosimetry
dose, drug infusion to light initiation interval (DLI) (Allison and Sibata 2008).
and light intensity as a crude but reproducible means Pulmonary lesions may also auto-fluoresce based
to achieve dosimetric success. Each PS will have a upon inherent chromophores in tumors (Dooms et al.
unique and optimal drug dose, DLI and intensity of 2010). Commercially available auto-fluorescent
light for activation. Until light dosimetry is better bronchoscopy units have already shown tremendous
defined and measured, PDT will not be able to value in lesion detection as well as during surveil-
achieve its full potential. lance bronchoscopy of high-risk populations. The use
Clinically, patients metabolize PS on an individu- of auto-fluorescence, PS fluorescence and fluorescent
alized basis. So when using a standard drug dose, changes post PDT is an area of active research.
light dose and DLI, some patients will have more
intense PDT than others due to what appears to be a
genetic characteristic of metabolism and, perhaps, 8 Rationale for Pulmonary PDT
immune response. For example, some of these indi-
viduals retain more PS drug both in tumor and normal PDT is one of many interventions available for pul-
tissue creating a very brisk PDR. monary lesions (Moghissi and Dixon 2003). PDT can
Currently the concept of photo bleaching is used to be employed simultaneously or sequentially with
assist in PDT dosimetry. As PS accumulates to a external beam radiation, brachytherapy, stenting,
greater degree in malignancy than normal tissue the YAG laser, cryotherapy, chemotherapy or surgery.
current guidelines are to employ a clinically deter- The versatility of PDT is one of this therapy’s
mined amount of PS that floods the tumor but stronger points. For select in situ or early invasive
attempts to stay below the threshold of a clinically lesions PDT alone can be curative. Bulky obstructing
significant PDR in surrounding normal tissue. Thus endobronchial lesions can be palliated. PDT can be
the PS in normal tissue will be used up (photo- used to downstage cancers for resection or as an
bleached) but not generate significant or permanent adjunct for tight or positives margins post-operatively
toxicity. If too much PS is infused the malignant or intra-operatively.
region will still retain extra PS, but the surrounding In general PDT is used as part of a multi-disci-
normal tissue will also have enough PS to generate a plinary approach for pulmonary disease control either
PDR which could be morbid. Ideally the optimal drug in a curative or palliative settings. Unlike other
dose and illumination dose would be individualized interventions PDT can be repeated to allow for
PDT-Lung 375

Fig. 2 Obstructing lesion


(left) of airway treated by
photofrin PDT, 2 mg/kg,
200 J Red light. Two weeks
post PDT (right) shows com-
plete response. (Photo’s cour-
tesy of Gordon Downie)

prolonged disease control. Interestingly PDT appears


to work regardless of histology as long as PS can be
activated by appropriate light source. One important
point is that following illumination PDT can take
hours or days to generate a clinical response so that it
is not indicated in the emergent setting. Further, blood
will preferentially absorb red light so hemostasis is
required for successful PDT. Also of importance is
that for photofrinÒ one must generally wait 48 h after
infusion to achieve a selective PDR. This shows the
need for other intervention in the emergent pulmonary
setting.

9 PDT Procedure

The actual PDT procedure for pulmonary disease has


evolved to the point that it can reproducibly ablate
Fig. 3 Fiber optic in place and illuminating tumor bed
lesions without undue acute or chronic mobility
(Moghissi and Dixon 2008). Following informed
consent the PS is introduced intravenously and the fiber optic through the bronchoscopes biopsy
allowed time to concentrate in the lesion. Endobron- channel. Various diffusing fibers of lengths from 1 to
chial tumors are generally approached endobronchi- 5 cm can be employed for illumination. For longer
ally. This can be via rigid or fiber optic bronchoscope. lesions the illuminating fiber can be stepped to ensure
Thoracic surgeons often use general anesthesia and total lesion illumination. For bulky tumors ([1 cm
apply the rigid bronchoscope to visualize central Deep) the light source may be inserted into the mass
lesions. Pulmonologists use conscious sedation and either mechanically or often after YAG laser or
apply fiber optic scopes to visualize lesions. Fluo- equivalent tunneling procedures. A typical PDT out-
rescent technology such as the life scope or other come is shown in Figs. 2 and 3. The PDT procedure
similar devices can be employed to enhance lesion can be offered in stented regions as well. Self-
visualization. Upon lesion visualization, the fiber expanding stents are not injured by the non-thermal
optic for treatment connected to a light source light treatment of PDT. Opaque stents will diminish
designed to activate that particular PS is brought to light transmittance so additional treatment time can
the lesion. This is generally accomplished by passing be required in this instance. Again, PDT can be
376 R. R. Allison

delivered in previously irradiated regions either from Current light sources produce intense illumination.
external beam or brachytherapy. In contrast to other During treatment both patients and practitioners need
endobronchial procedures PDT can be directly to wear eye protecting goggles. Eye injury or blind-
observed which ensures accurate targeting. At the ness can result from improper safety procedures.
completion of the PDT session tumors appear dusky
and hypoxic. At 48 h (or sooner) post PDT, a second
bronchoscopy session is always undertaken to remove 11 Clinical Outcomes
necrotic debris and if viable tumor is seen an addi-
tional PDT session is possible when photofrinÒ is the Among the first clinical success for PDT was the
PS. Necrotic tumor can be brought out in bulk via treatment of pulmonary lesions (Kato et al. 1996).
cryotherapy which may also assist in improving Early studies included mostly central endobronchial
tumor control. It is critically important to always advanced cases that had failed radiation therapy and
include follow up bronchoscopy post PDT to ensure other intervention such as YAG Laser or electrocau-
adequate pulmonary toilet. Failure to do so can result tery. Pulmonary palliation was generally achieved
in fatal airway obstruction, fortunately a quite rare showing not only PDT’s ability as salvage therapy but
consequence. also its relative safety in combination with other
More peripheral lesions can be treated via trans- treatments. In a more recent review it appears that
bonchial application of the light source using endo- intentionally offering multi-modal salvage allows for
bronchial navigational guidance. These navigational prolonged survival and palliation for locally advanced
tools have opened new pathways for PDT to more endobronchial disease (Santos et al. 2004).
peripheral lesions. CT-guided placement of light Currently PDT has focused on the treatment of
sources is also possible for peripheral lesions as is a early lesions. In combination with fluorescence
video-assisted thoracoscopic approach (Moghissi detection, high rates of lesion ablation with preser-
et al. 2003). An open surgical procedure with lesion vation of pulmonary function are routinely achieved
resection or debulking followed by planned PDT is (Weigel et al. 1999). Select publications from the
also an option for nodules and diffuse tumors such as PDT literature follow.
mesothelioma. For diffuse lesions multiple light
sources are required to minimize treatment times.
11.1 Obstructing Endobrochial Lesions

10 PDT-Morbidity These highly symptomatic tumors lead to respiratory


compromise, hemoptysis and infection and can be
Morbidity can occur immediately following PS infu- fatal. Local palliation improves quality of life and can
sion as the individual becomes photosensitive within prolong survival. In an early pivotal study Moghissi
minutes of infusion. Individuals who will not follow et al. (1993) reported a randomized trial comparing
sunlight precautions should not undergo PDT. Photo- laser ablation to PDT for symptomatic central endo-
sensitivity to sunlight exposed anatomy can occur even bronchial tumors. Response rates leading to pulmon-
with only a few minutes of sunlight exposure. Clini- ary palliation were equivalent and approached 80%,
cally this is treated as one would a moderate to severe however, PDT offered a statistically significant and
sunburn, generally using steroids and pain control. meaningful prolongation of palliation measured in
PDT treatment is non-thermal and does not have a months. Similarly a randomized trial comparing pal-
significant rate of added acute morbidity compared to liative radiation therapy (30 Gray/10fx) to radiation
bronchoscopy alone or when employed with other therapy (30 Gray/10fx) and PDT reported superior
endobronchial or surgical approaches. With the rou- pulmonary palliation with the addition of PDT again
tine addition of follow-up bronchoscopy emergent measured in months (Lam et al. 1987).
airway obstruction is virtually non-existent. Even In a report by Minnich et al. (2010) on PDT to 133
when combined with HDR Brachytherapy, morbidity patients, mainly with obstructing lesions of the main
appears limited, and generally mild, except in a very stem bronchus, one or two PDT sessions allowed for a
rare case of fistula (Sanfilippo et al. 2001). clinically significant reduction in dyspnea as well as
PDT-Lung 377

prolonged pulmonary palliation. Only four patients commercially available in Japan. Kato, in a series of
reported a photosensitivity reaction. Of note is that reports (Furukawa et al. 2005; Kato et al. 2003; Usuda
patients in this series had failed prior endobronchial et al. 2007), showed a 92% complete response with
therapies. this drug. Interestingly this group employs fluores-
Similarly in a report of 100 cases of PDT to cence and auto-fluorescence to improve targeting and
patients with advanced unresectible lung cancer to monitor treatment outcome. With its short period of
(Moghissi et al. 1999), PDT achieved excellent pul- cutaneous photosensitivity MACE has become the
monary palliation. Two-year survival was 20% preferred PS for pulmonary disease in Japan.
revealing the ability of PDT to offer very prolonged
palliation in select cases. A number of other studies
show excellent salvage rates of 80% or more with 11.3 Multiple Primary Lung Cancer
PDT alone (Moghissi and Dixon 2003).
In general these individuals are heavy smokers with
limited lung function. Treatment options are limited
11.2 Early Lung Cancer but PDT has found a niche here. In a study from Japan,
patients with multiple primary lung cancers underwent
This is a heterogeneous group of lesions varying from either PDT alone or PDT to some lesions and surgery to
in situ to minimally invasive disease. Invasive lesions other lesions. PDT alone or in conjunction with surgery
can spread to regional lymph nodes, and as these allowed for 100% CR with all patients alive and
nodes cannot be treated by PDT, patient selection is breathing well (Usuda et al. 2010). An aggressive sur-
critical. Ultrasound, to assist in-depth invasion, can be gical approach of pneumonectomy or lobectomy and
of critical utility (Miyazu et al. 2002). In contrast to PDT was presented by Jung et al (Jung et al. 2010).
resection PDT will maintain the pulmonary paren- Post-operative mortality was 9%. The authors felt that
chyma, an attractive reason to employ this modality. PDT alone or in combination with limited resection
Further, patients medically inoperable or who refuse could achieve excellent outcome, perhaps avoiding the
surgery can still undergo therapy to prevent disease mortality associated with the larger surgery. In a study
progression. In a study from the Mayo clinic one or from Russia, multiple lesions smaller than 1 cm could
two PDT sessions eliminated early invasive or in situ be routinely ablated by PDT and was the treatment of
disease in 92% of patients (Cortese et al. 1998). choice for early synchronous or metachronous multiple
Similar excellent outcomes has been noted by other primary lung cancers (Sokolov et al. 2010).
investigators. Moghissi et al. (2007), with several year
follow-up, reported that one or rarely two PDT ses-
sions allowed a 100% pathologic complete response 11.4 Down Staging
for lesions centrally located. No patient had pul-
monary compromise and only one had sunlight sen- As surgical resection remains the best chance for cure
sitivity. Furuse et al. (1993) achieved an 81% CR rate for advanced lung cancer PDT has been examined in a
with a single PDT session and a second session was neo-adjuvant role for tumor debulking to allow for
able to salvage most failures. Of note in this series of excision. In a study of 41 patents, Ross et al. (2006)
51 patients, sunburn from photosensitivity reaction reported that half the patients were down staged enough
was noted in 25%, showing the need for sunlight by PDT for curative resection. A small cohort had
exposure precautions. Furukawa et al. (2005), in a complete pathologic response to pre-operative PDT
series of 114 patients, achieved a 93% CR for lesions treatment. Pre-operative PDT also allowed for com-
up to 1 cm but only a 60% CR for larger lesions. The plete resection in a report by Mortman and Frankel
larger lesions could often be salvaged by multiple (2006) as well as DeArmond et al. (2008). PDT allowed
PDT session or other interventions. This proves that for resection in 22 of 26 cases reported by Okunaka
in bulky lesions, single session PDT treatment is Tetsuya et al. (1999). In this study a statistically
limited to illumination depth of about 1 cm. More significant increase in survival was also noted for T3
recently, the use of MACE, rather than photofrin, as a lesions (Main Bronchus) when comparing pre-
PS has been reported. MACE is on label and operative PDT to no PDT for this cohort of patients.
378 R. R. Allison

11.5 Peripheral Lesions patients in the radiation + PDT cohort achieved


complete remission which offered prolonged survival.
With technologic advances peripheral lesions have PDT has also been intentionally combined with
become amenable to PDT. These tumors can be HDR Brachytherapy. In a study by Freitag et al.
approached in various ways including video-assisted (2004), 32 patients with bulky endobronchial non-
thorascopy (Moghissi et al. 2003), CT-guided place- small-cell lung cancer underwent photofrin PDT
ment of illumination fibers and via bronchoscopy followed by five fractions of HDR (4 Gray delivered
augmented with guidance. Using CT guidance, cath- to 1 cm depth). HDR was delivered weekly starting
eters were placed in peripheral tumors and then fiber on week six post PDT. At 2-year follow-up 26
optics introduced for illumination. As reported by patients were NED (81%). All these individuals were
Okunaka et al. (2004) high tumor response rates were salvaged by additional PDT, HDR or external beam
noted but as might be expected, a 20% pneumothorax radiation. No severe complications or hospitalization
rate was also seen. Moghissi et al. (2003) described a were seen. In particular no fistula or hemoptysis was
direct visualization procedure via VATS that also reported. Similar outcomes were reported by
achieved high response without pneumothorax. Weinberg et al. (2010). In this series of nine cases,
HDR was generally the initial therapy with three once
weekly HDR sessions delivering 5 Gray to 5 mm
11.6 Pleural Tumors followed 1 month later by photofrin PDT. A complete
response rate of 80% was noted with all patients who
Mesothelioma and advanced non-small-cell cancers achieved CR remaining NED for up to 5 years. Two
can progress plurally leading to demise. Current cases of mild sunlight photosensitivity were seen.
options are limited and PDT has been explored for
their indication. In preliminary studies the combina-
tion of maximal resection followed by PDT to the
12 Conclusion
involved anatomy was undertaken. (Friedberg et al.
2004; Du et al. 2010). Multiple light sources are
Photodynamic therapy has a long and successful track
required for this extensive volume of illumination. In
record in the treatment of pulmonary malignancies. PDT
select cases radiation has also been added to the
can ablate a wide variety of histologies as long as the
treatment mix. While preliminary, high-response rates
drug can be infused and light sources can be brought to
are possible but due to technical difficulties morbidity
the lesion. Of critical importance is that time is required
occurs. Further study is warranted.
for lesion response so that PDT should not be employed
up front in situations where pulmonary compromise
exists. The great strength of PDT is that it can be
11.7 PDT in Combination with Radiation
employed simultaneously or serially with other estab-
lished oncologic interventions to assist in disease control
As mentioned, early PDT trials included patients who
in thoracic cancer. PDT fits in well with the concept of a
had failed other interventions, particularly radiation
multi-disciplinary approach to thoracic oncology.
therapy. High-response rates without undue morbidly
Viewing PDT as complimentary rather than competitive
were seen. One of the few randomized trials in pul-
ensures the patient the best chance of clinical benefit.
monary PDT explored the intentional combination of
external beam therapy with PhotofrinÒ PDT (Lam
et al. 1987). Patients with histologically proven, un-
resectable, obstructive, non-small-cell bronchogenic References
carcinoma were randomized to radiation therapy
alone or radiation therapy followed by PDT. While Allison RR, Sibaba CH (2010) Oncologic photodynamic
the size of the study was small, 80% of radiation therapy photo sensitizers: a clinical review. Photodiagnosis
Photodyn Ther 7:61–75
therapy alone patients had symptomatically pro- Allison RR, Sibata CH (2008) Photodiagnosis for cutaneous
gressed by 12 week post therapy, compared to only malignancy: a brief clinical and technical review. Photo-
20% in the PDT + radiation group. Further only diagnosis Photodyn Ther 5:247–250
PDT-Lung 379

Allison RR, Mota HC, Sibata CH (2004a) Clinical PD/PDT in using Photofrin II combined with palliative radiotherapy
North America: An Historical Review. Photodiagnosis versus palliative radiotherapy alone in patients with inop-
Photodyn Ther 1:263–277 erable obstructive non-small cell bronchogenic carcinoma.
Allison RR, Downie GH, Cuenca R, Hu XH, Childs CJH, Photochem Photobiol 46:893–897
Sibata CH (2004b) Photosensitizers in clinical PDT. Mang TS, Dougherty TJ, Potter WR, Botle DG, Somer S,
Photodiagnosis Photodyn Ther 1:27–42 Moan J (1987) Photobleaching of poryrins used in photo-
Cortese DA, Edell ES, Kinsey JH (1998) Photodynamic therapy dynamic therapy and implications for therapy. Photochem
for early stage squamous cell carcinoma of the lung. Mayo Photobiol 45:501–506
Clinic Proc 72:595–602 Minnich DJ, Bryant AS, Dooley A, Cerfolio RJ (2010)
Daniell MD, Hill JS (1991) A history of photodynamic therapy. Photodynamic laser therapy for lesions in the airway. Ann
Aust N Z J Surg 61:340–348 Thorac Surg 89:1744–1748
DeArmond DT, Mahtabifard A, Fuller CB, McKenna RJ Jr Miyazu Y, Miyazawa T, Kurimoto N, Iwamoto Y, Kanoh K,
(2008) Photodynamic therapy followed by thoracoscopic Kohno N (2002) Endobronchial ultrasonography in the
sleeve lobectomy for locally advanced lung cancer. Ann assessment of centrally located early stage lung cancer before
Thorac Surg 85:e24–e26 photodynamic therapy. Am J Respir Crit Care Med 165:832
Dooms C, Seijo L, Gasparini S, Trisolini R, Ninane V, Moghissi K, Dixon K, Parson RJ (1993) A controlled trial of
Tournoy KG (2010) Diagnostic bronchoscopy: state of the ND-YAG lasers vs photodynamic therapy for advanced
art. Eur Respir Rev 19:229–236 malignant bronchial obstruction. Lasers Med Sci 8:269–273
Dougherty TJ (1996) A brief history of clinical photodynamic Moghissi K, Dixon K (2003) Is bronchoscopic photodynamic
therapy development at Rosewell Park Cancer Institute. therapy a therapeutic option in lung cancer? Eur Respir J
J Clin Laser Med Surg 14:219–221 22:535–541
Dougherty TJ, Marcus SL (1992) Photodynamic therapy. Eur J Moghissi K, Dixon K (2008) Update on the current indications,
Cancer 10:1734–1742 practice and results of photodynamic therapy (PDT) in early
Du KL, Both S, Friedberg JS, Rengan R, Hahn SM, Cengel KA central lung cancer (ECLC). Photodiagnosis Photodyn Ther
(2010) Extrapleural pneumonectomy, photodynamic ther- 5:10–18
apy and intensity modulated radiation therapy for the Moghissi K, Dixon K, Stringer M, Freeman T, Thorpe A,
treatment of malignant pleural mesothelioma. Cancer Biol Brown S (1999) The place of bronchoscopic photodynamic
Ther 10:425–429 therapy in advanced unresectable lung cancer: experience of
Freitag L, Ernst A, Thomas M, Prenzel R, Wahlers B, Macha HN 100 cases. Eur J Cardiothorac Surg 15:1–6
(2004) Sequential photodynamic therapy (PDT) and high dose Moghissi K, Dixon K, Thorpe JA (2003) A method for video-
brachytherapy for endobronchialtumour control in patients assisted thoracoscopic photodynamic therapy (VAT-PDT).
with limited bronchogenic carcinoma. Thorax 59:790–793 Interact Cardiovasc Thorac Surg 2:373–375
Friedberg JS, Mick R, Stevenson JP, Zhu T, Busch TM, Shin D, Moghissi K, Dixon K, Thorpe JAC, Stringer MR (2007)
Smith D, Culligan M, Diofte A, Clatstein E, Hahn SM Photodynamic theray in early central lung cancer: a
(2004) Phase II trial of pleural photodynamic therapy and treatment option for patients ineligible for surgical resec-
surgery for patients with non-small-cell lung cancer with tion. Thorax 65:391–395
pleural spread. J Clin Oncol 22:2192–2201 Mortman KD, Frankel KM (2006) Pulmonary resection after
Furukawa K, Kato H, Konaka C, Okunaka T, Usada J, successful downstaging with photodynamic therapy. Ann
Ebihara Y (2005) Locally recurrent central type early stage Thorac Surg 82:722–724
lung cancer \1.0 cm in diameter after complete remission Okunaka T, Kato H, Tsutsui H, Ishizumi T, Ichinose S,
by Photodynamic Therapy. Chest 128:3264–3275 Kuroiwa Y (2004) Photodynamic therapy for peripheral
Furuse K, Fukuoka M, Kato H et al (1993) A prospective Phase lung cancer. Lung Cancer 43:77–82
II stude on Photodynamic Therapy with Photofrin II for Okunaka T, Hyoshi T, Furukawa K, Yamamoto H, Tsuchida T,
centrally located early stage lunch cancer. J Clin Oncol Usuda J, Kumasaka H, Ishida J, Konaka C, Kato H (1999)
11:1852–1857 Lung cancer treated with photodynamic therapy and
Jung EJ, Lee JH, Jeon K, Koh WJ, Suh GY, Chung MP, Kim H, surgery. Diagn Ther Endosc 5:155–160
Kwon OJ, Shim YM, Um SW (2010) Treatment outcomes Raab O (1900) On the effect of fluorescent substances on
for patients with synchronous multiple primary non-small infusoria (German). Z Bilogoy 39:524
cell lung cancer. Lung Cancer, Dec 8, Epub ahead of print Ross P Jr, Grecula J, Bekaii-Saab T, Villalona-Calero M,
Kato H, Okunaka T, Shimatani H (1996) Photodynamic therapy Otterson G, Magro C (2006) Incroporation of photodynamic
for early stage bronchogenic carcinoma. J Clin Laser Med therapy as an induction modality in non-small cell lung
Surg 14:235–238 cancer. Lasers Surg Med 38:881–889
Kato H, Furukawa K, Sato M, Okunaka T, Kusunoki Y, Sanfilippo NJ, HIS A, DeNittis AS, Ginsberg GG,
Kawahara M, Fukuoka M, Miyazawa T, Yana T, Matsui K, Kochman ML, Friedberg JS, Hahn SM (2001) Toxicity of
Shiraishi T, Horinouchi H (2003) Phase II clinical study of photodynamic therapy after combined external beam radio-
photodynamic therapy using mono-L-aspartylchlorin e6 and therapy and intraluminal brachytherapy for carcinoma of the
diode laser for early superficial squamous cell carcinoma of upper areodigestive tract. Lasers Surg Med 28:278–281
the lung. Lung Cancer 42:103–111 Santos RS, Radtopoulos Y, Keenan RJ, Jalal A, Maley RH,
Lam S, Kostashuk EC, Coy EP, Laukkanen E, LeRiche JC, Landreneau RJ (2004) Bronchoscopic palliation of primary
Meuller HA, Szasz IJ (1987) A randomized comparative lung cancer: single or multimodality therapy? Surg Endosc
study of the safety and efficacy of photodynamic therapy 18:9316
380 R. R. Allison

Sibata CH, Colussi VC, Oleinick NL, Kinsella TJ (2000) in patients with centrally located early cancer lesions. J Thorac
Photodynamic therapy: a new concept in medical treatment. Oncol 5:62–68
Braz J Med Biol Res 33:869–880 Von Tappeiner H, Jesionek A (1907) The sensitizing action of
Sokolov VV, Telegina LV, Trakhtenberg AKH, Kolbanov KI, fluorescent substances. An overall account of investigations
Pikin OV, Frank GA (2010) Enodobronchial surgery and on photodynamic phenomena. Leipzig, F.C.W. Vogel
photodynamic therapy for the treatment of multiple primary Von Tappeiner H, Jesionek A (1903) TherapeutischeVersu-
lung cancer. Khirurhiia 7:28–31 chemitfloreszierendenStofffen (Therapeutic experiments
Usuda J, Tsutsui H, Honda H, Ichinose S, Ishizumi T, Hirata T, with fluorescent substances). Muench Med Wochenscr
Inoue T, Ohtani K, Maehara S, Imai K, Tsunoda Y, 47:2042–2044
Kubota M, Ikeda N, Furukawa K, Okunaka T, Kato H Weigel TL, Kosco PJ, Dacic S, Yousem S, Luketich JD (1999)
(2007) Photodynamic therapy for lung cancers based on Fluorescence bronchoscopic surveillance in patients with a
novel photodynamic diagnosis using talaporfin sodium history of non-small cell lung cancer. Diagn Ther Endosc 6:1–7
(NPe6) and autofluorescence bronchoscopy. Lung Cancer Weinberg BD, Allison RR, Sibata C, Parent T, Downie G
58:317–323 (2010) Results of combined photodynamic therapy (PDT)
Usuda J, Ichinose S, Ishizumi T, Hayashi H, Ohtani K, Maehara S, and high dose brachytherapy (HDR) in treatment of
Ono S, Kajiwara N, Uchida O, Tsutsui H, Ohira T, Kato H, obstructive endobronchial non-small cell lung cancer
Ikeda N (2010) Management of multiple primary lung cancer (NSCLC). Photodiagnosis Photodyn Ther 7:50–58
The Role of Radiofrequency Ablation
in the Treatment of Stage 1 Non-Small
Cell Lung Cancer
John M. Varlotto, Julia A. Shelkey, and Rickhesvar P. Mahraj

Contents Abstract
Currently, the standard of care for stage I non-small
1 Introduction.............................................................. 382 cell lung cancer (NSCLC) is surgical resection.
2 Patient Selection and Evaluation........................... 382 Although this treatment modality has been demon-
strated to have 5 year survival rates approaching
3 Technique.................................................................. 383
80%, there need to be effective alternative treat-
4 Results ....................................................................... 385 ments for patients who are medically inoperable.
4.1 Clinical Results.......................................................... 385
Radiofrequency ablation (RFA) has emerged as a
4.2 Ablate and Resect Studies......................................... 390
minimally-invasive therapy to fill this void. This
5 Follow-Up.................................................................. 391 modality has been found to be most effective for
6 Summary................................................................... 393 treatment of small (\3 cm), peripheral lesions that
References.......................................................................... 394 are located distal to vasculature, large airways, and
the mediastinum. The most common complications
after RFA include pneumothorax, pneumonia, and
pleural effusion. To date, accurate assessment of the
efficacy of RFA has been difficult to determine due
to short follow-up times of current studies and the
lack of standard definitions of local recurrence as
well as toxicity. Current literature has suggested
local progression rates ranging from 20 to 42%, but
assessment by prospective trials with long-term
follow-up and standardized definitions of toxicity
and local control are needed to determine the true
benefit of this procedure.

J. M. Varlotto (&)  J. A. Shelkey


Radiation Oncology—CH63,
Penn State Hershey Cancer Institute,
500 University Drive, Abbreviations
PO Box 850, Hershey,
PA 17033, USA
AICD Automatic implantable cardioverter
e-mail: jvarlotto@hmc.psu.edu defibrillator
R. P. Mahraj
AZ Ablation zone
Department of Radiology, C Centigrade
Penn State Hershey Medical Center, CA Complete ablation
500 University Drive, Hershey, CN Complete necrosis
PA 17033, USA

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 381
DOI: 10.1007/174_2011_230, Ó Springer-Verlag Berlin Heidelberg 2011
382 J. M. Varlotto et al.

CR Complete response concerning percutaneous RFA of lung neoplasms in


CSS Cancer specific survival the recent literature. This technique directly destroys
CTCAE Common terminology criteria for tumor cells by delivering a high-frequency, alternat-
adverse events ing current with frictional heating and ionic agitation.
DFS Disease-free survival Consequently, cell membrane alteration, protein
DR Distal recurrence denaturation, and necrosis around the electrode are
EBUS Endobronchial ultrasound directly produced. However, this process can indi-
EP Extrapulmonary recurrence rectly stimulate the immune system because it relea-
F/U Follow-up ses large amounts of tumor antigen and tissue debris
GGA Ground-glass attenuation into the systemic circulation (Fietta et al. 2009;
H&E Hematoxylin and eosin staining Widenmeyer et al. 2010). The treatment of lung
H&M nodes Hilar and mediastinal nodes neoplasms may be ideal for RFA because the sur-
IA Incomplete ablation rounding pulmonary tissue may cause an insulating
IP Intrapulmonary recurrence effect whereby the heat is concentrated within the
LC Local control tumor. The low thermal conductivity of the neigh-
LR Local recurrence boring lung tissue has been demonstrated to greatly
MAM Monoclonal anti-mitochondrial increase radiofrequency-induced temperatures within
antibodies a defined ablation target (Liu et al. 2006).
Mn Months The purpose of this chapter is to describe the
NS Not stated technique, results, complications, and follow-up
NSCLC Non-small cell lung cancer evaluation of Stage I Non-small cell lung cancer
PFS Progression-free survival (NSCLC) patients treated with RFA.
PNB Percutaneous biopsy
Pneumo Pneumothorax
R Recurrence 2 Patient Selection and Evaluation
RECIST Response evaluation criteria in solid
tumors The treatment approach for effective management of
RF Radiofrequency early stage NSCLC is evolving to mirror complex dis-
RFA Radiofrequency ablation ease presentations and patient situations, as well as to
RT External beam radiotherapy accommodate the many different therapeutic options.
RT-br Brachytherapy For early stage lung cancer, surgery offers survival rates
RTOG Radiation therapy oncology group as high as 79.5% at 5 years (Koike et al. 1998). Many
S Survival patients, however, are inoperable due to either co-mor-
SBRT Stereotactic body radiation therapy bidities or the development of new or recurrent lung
SVB Supravital blue staining cancer following past therapeutic intervention. Under
these circumstances, radiofrequency ablation is a mini-
mally-invasive technique that represents an additional
tool to treat patients in these situations.
The decision to offer this method of treatment is best
1 Introduction considered in a multi-disciplinary group setting. Tho-
racic surgeons, radiation oncologists, thoracic inter-
Interstitial hyperthermia via the radiofrequency abla- ventional radiologists, and medical oncologists should
tion of a primary lung cancer was first described be involved in determining the most appropriate treat-
during an open thoracotomy in 1983 (Lilly et al. ment strategy. This plan should include evaluation of
1983). The first report of modern, percutaneous surgical technique (lobectomy versus limited resec-
radiofrequency ablation in patients with lung malig- tion), conventional radiotherapy versus SBRT, and
nancies was published in 2000 (Dupuy et al. 2000). RFA or combined modality treatment. RFA can be
Due to the technique’s minimal invasiveness, there considered as a sole treatment option or with other
has been considerable interest and multiple case series local modalities (external beam radiotherapy, SBRT,
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer 383

brachytherapy) to achieve local tumor obliteration with which causes ions within the tumor to oscillate lead-
the goal of cure or palliation. ing to frictional heat. The area of coagulation necrosis
Absolute contraindications exist, including severe achieved is related to the strength of the radiofre-
COPD with FEV1 \0.5 L, severe pulmonary artery quency energy, the current-carrying time, the diame-
hypertension, co-existent pneumonia, past pneumo- ter and shape of the electrode, and the composition of
nectomy, and uncorrectable coagulopathy. Further- the surrounding tissues. The lung may be particularly
more, the treatment of lesions located centrally next suited for this technique due to the insulating effect
to large vessels must be individualized due to the provided by the surrounding, air-filled, pulmonary
higher risks of ineffective ablation and complications. parenchyma, which improves the effectiveness of
Patients with automatic implantable cardioverter RFA by concentrating the heat energy in the tumor
defibrillators (AICDs) and pacemaker devices repre- (White and D’Amico 2008). Cellular death is induced
sent a special risk and require expert electrophysio- by thermal coagulation necrosis at an optimal heating
logical management. Stereotactic body radiation temperature of 60–1058C (Matsuoka and Okuma
therapy (SBRT), microwave, and cryoablation repre- 2007). Above this temperature, carbonization and gas
sent alternative ablation techniques for such patients. formation occur, which in turn lead to an increase in
As with selection of patient for other curative tissue impedance and restriction in frictional heating
treatments, a thorough diagnosis and staging should of the tumor tissue (White and D’Amico 2008).
be preformed. At our institution, FDG-PET as well as There are three systems approved by the FDA to
percutaneous biopsy (PNB) of extrathoracic lesions as ablate soft tissue tumors that can also be used to
necessary are part of the standard pre-treatment therapeutically destroy lung tumors. They all have an
staging. Nodal staging is preferred. A bronchoscopy automatic feedback of either temperature (RITA
with nodal sampling via endobronchial ultrasound Medical Systems Freemont, CA) or a specific change
(EBUS) is considered. If nodal sampling via EBUS is in impedance (Valleylab: Boulder, CO; and Boston
negative and a patient can tolerate general anesthesia Scientific LeVeen CoAccessTM Electrode System,
a mediastinoscopy is performed. As per past guide- Natick, MA). The authors’ experience is with the
lines, if the staging work-up with FDG CT/PET is Valleylab, cool-tip system and RITA Medical
negative, patients with clinical T1 tumors in the outer Systems. The cool-tip system is preferred for small
2/3 of the hemithorax can forego invasive nodal lesions under 2 cm, while the RITA system is used for
sampling (Detterbeck et al. 2007). large lesions of 2–3 cm. The Valleylab, cool-tip sys-
tem causes less collateral damage to normal tissues
and prevents tissue charring (excessive tissue
3 Technique destructive near the electrode resulting in less effec-
tive treatment of distal tumor tissue) due to its use of a
The procedure can be performed under conscious single small gauge needle and because of a continu-
sedation or general anesthesia. General anesthesia is ous infusion of iced-water around the probe as the
usually reserved for extreme pain, lack of patient tumor is heated resulting in improved radiofrequency
co-operation, and necessity for airway control during (RF) energy distribution and increased ablation
suspended respiration. If bleeding is a significant risk, diameter (Casal et al. 2010) (Fig. 1a, b). The different
double-lumen endotracheal tubes are recommended. needle types are selected based on tumor size and
Most patients who are able to tolerate a CT-guided location, as well as preference and availability.
needle biopsy of the lung are generally candidates for Needle size generally ranges from 14 to 17 gauge.
RFA (McTaggart and Dupuy 2007). Larger tumors ([3 cm) are also associated with worse
RFA utilizes a radiofrequency generator to supply local control via RFA (Lanuti et al. 2009; Lee et al.
power through an electrode. The electrode is in the 2003). However, larger electrodes can produce
shape of needle, which is placed under CT-fluoro- necrosis measuring up to 4–5 cm in diameter. This
scopic guidance into the lesion. The patient essen- allows for the treatment of a 3 cm lesion with a 1 cm
tially becomes part of an electrical circuit in which a margin of normal lung. Preferentially, when available,
radiofrequency generator produces an alternating a multiple tine system is used to heat larger lesions
electrical field. Heat is located within the needle, and reduce procedure times because of its ability to
384 J. M. Varlotto et al.

in continuity with large blood vessels may be sub-


optimally treated with RFA.
CT volumetric scanning with multi-planar recon-
struction are effective methods to evaluate and guide
needle placement within the tumor. RFA is ideally
performed under CT-fluoroscopic guidance, which
allows real-time needle guidance for speed and
accuracy. Probe placement depends on the size of the
lesion and the thermal ablation volume characteristics
of the needle. A single needle may be re-inserted into
the lesion multiple times to achieve overlapping
volumes that can completely ablate the tumor. Mul-
tiple placements are particularly helpful for irregular
lesions. The average time for a single 2–3 cm volume
ablation is 12 min.
Assessment of adequate thermal ablation of the
tumor requires demonstrating a minimal core tumor
temperature of 608C and evaluating the thermal injury
occurring to the normal lung surrounding the tumor.
The latter has been shown using porcine lung models
in which the ground-glass attenuation (GGA) CT scan
changes taking place in the lung correspond to hem-
orrhage that occurs beyond the volume of tissue
necrosis produced by thermal ablation (Yamamoto
et al. 2005). Knowledge of pre-ablation lesion size is
important so that thermal injury can be demonstrated
beyond the margin of the lesion. This assessment can
be made complex due to procedure-related enlarge-
ment of the solid portion of the tumor and the syn-
chronous development of the peripheral zone of
GGA. As a guideline, 0.5–1 cm of circumferential
Fig. 1 a Pathologic specimen demonstrating complete abla- GGA surrounding the lesion indicates a satisfactory
tion due to effective heating throughout the tumor. b Pathologic treatment endpoint. Additional ablation with reposi-
specimen that demonstrates charring with viable tumor within tioning of the electrode needle should be considered
the treatment zone. Charring prevents homogeneus thermal
energy transfer throughout tumor, resulting in ineffective to treat areas where GGA is absent (Fig. 3a, b). One
treatment in tumor sites distal to the RFA probe group of investigators, however, cautioned that the
extent of the GGA may not be indicative of the exact
extent of coagulation necrosis (Hiraki et al. 2007).
Post-procedure recovery follows conscious seda-
open like the spokes of an umbrella. For peripheral tion or post-anesthesia recovery standards. The lung
lesions, pleuritis and rib pain can be avoided by puncture site should be kept dependent for 2–3 h.
deliberately inducing a small pneumothorax during Antitussive medications may be beneficial and ade-
the procedure (Fig. 2a, b). quate pain control as well as monitoring of vital signs is
A particular problem is encountered with large necessary. Chest X-rays should be taken at 1 and 4 h
blood vessels ([3 mm) near a tumor. The vasculature following the procedure to exclude pneumothorax or
constantly cools the heated tissue because the flowing occult pulmonary hemorrhage. Patients may be dis-
blood causes conduction loss from the area being charged 6–8 h the same day or observed overnight as
treated. This difficulty is commonly known as the indicated. A mild fever is common for the initial 2 days
heat-sink effect (Rose et al. 2006). As a result, tumors following the procedure (Yamamoto et al. 2005).
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer 385

Fig. 2 a Peripheral-based
tumor presenting for RFA.
b A controlled pneumothorax
(injection of 250 cc of air in
this example) was used to
separate the tumor from the
chest wall in order to prevent
complications including
pleuritis, neuritis, and bone
pain

Fig. 3 a Thermal ablation of a


2.4 cm NSCLC with absence
of GGA noted anteriorly
indicating incomplete tumor
necrosis. b Effective ablation
achieved radiographically after
repositioning of the electrode
anteriorly

an ultrasound-guided technique. Hiraki et al. (2007)


4 Results were the only investigators who used CT-fluoroscopy,
which allows for real-time imaging at the expense of a
4.1 Clinical Results much higher procedure-related radiation exposure (Rose
2008). In general, the patients selected for RFA were
The results of RFA in the treatment of Stage I lung mostly medically-inoperable secondary to multiple
cancer are summarized in Table 1. Some series also medical co-morbidites with only a small minority
included metastatic tumors and/or more advanced lung refusing surgery. Patient follow-up in all series was
cancers, but the information in the tables contains only generally limited secondary to patient longevity.
details concerning stage I patients unless otherwise Tumors that were selected for treatment were generally
indicated. All patient series are retrospective single small and peripheral in location. Central tumors were
institution investigations, except the report by Lencioni avoided because of the heat-sink effect, complications
et al. (2008), which was a multi-institutional, prospec- due to proximity of nearby vital structures, and the
tive study. A percutaneous, CT-guided RFA technique necessity of longer needle trajectory through the aerated
was used in most studies. Ambrogi et al. (2007), how- lung (Pennathur et al. 2010; Steinke et al. 2005; Gilliams
ever, treated some tumors involving the chest wall with and Lees 2007; Lee et al. 2003). In most series, the aim of
Table 1 Summary of current clinical studies using RFA for treatment of Stage I lung cancer
386

Author #/size LR Definition F/U Local Control Complications Survival


Patient (mn)
Pop.
Lencioni* 13/ all \ 3.5 cm, Modified RECIST using CT Scan 1 month after 15* 92% CSS Major Complications- 2 yr S-75% and
2008 median 1.7* treatment as reference drainage needed* CSS-92%
1. Large or
symptomatic
pneumo-19.7%;
2. Pleural effusion-
NSCLC and CR = complete ablation of tumor lasting one year 2.9%
Lung
Metastases Minor complications-
self-limited
1. hemorrage-2.18%
2. pneumo-20.4%
3. effusion-8.02%
Hiraki T 20/ Mean tumor CR-No contrast enhancement in entire ablation zone, 21.8 72, 63, & 63% at 1, 2, Pneumothorax-57% S-90, 84, 74 at
2007 size = 2.4 or when the ablation zone exhibited a peripheral rim and 3 years 1, 2, and
(1.3-6 cm) of contrast-enhancement that was concentric, 3 years
symmetric and uniform with smooth margins.
IA-14 Pleural effusion-17%
Stage I Progression-circumferential enlargement. Irregular, Median time for local
NSCLC scattered, nodular, or eccentric foci in the AZ progression = 9 months One chest tube CSS-100, 93, 83
only IB-6 placement-rest at 1, 2, and
conservative 3 years
treatment
Pennathur 19/2.6 Modified RECIST-Based upon CT size and mass 29 42% local progression Pneumo-63%-pigtail S-95%-1 yr,
2007 (1.6-3.8 cm) quality; higher SUV or larger area on PET Scan catheter 68%-2 yr

Stage I Median time to local Prolonged air


NSCLC progression = 27 months leak [ 5 days-5%
only
Beland* 79/2.5 LC-lack of contrast enhancement in AZ 17 57%-no recurrence* NS Median DFS
2010 (1.5-5.5 cm) 38% LR 23 months*

NSCLC 19(24%)-RT Progression-focal enhancement [ 15 HU compared 18% IP


with unenhanced series
Different 9(11%) –RT-Br 18% Nodal
Stages
(continued)
J. M. Varlotto et al.
Table 1 (continued)
Author #/size LR Definition F/U Local Control Complications Survival
Patient (mn)
Pop.
94% Stage I Benign peritumoral enhancement-Symmetric rim of 6% Mixed
peripheral enhancement of \ 5 mm up to 6 mn after
ablation

7% Stage IIIB Growth on serial CT scans-used for patients intolerant 21% DR


of contrast
R related to size and
stage;
RT and RT-br
borderline
significance for R
Lanuti 2009 31pts/2.0 Lack of focal or diffuse enlargement of ablated lesion 17.3 31.5% LR Pneumo-13% S-2 & 4 yr-78%,
(0.8-4.4 cm) on CT and no evidence of eccentric enhancement on 47%
PET with minimum of 3 mn of FU;
Stage I
NSCLC only 3 with stable Progression of FDG H&M nodes after 3 mn-local
disease failure 50% LR with Pneumonia-16% 3 yr PFS and
received RT size [ 3 cm(3/6) DFS-58%, 39%
Effusion-21%

BP fistula-8%

Chest tube-8%

Transient recurrent
nerve palsy-1%
Rossi 2006 15 LUNG Increasing size or enhancement at tumor site-using 30 Day 11.4 mn 20% LR No major 9/15 alive
Ca/2.2 ct scan, repeat procedure-if 30 day positive, then mark as complications* without disease
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer

(1.0-3.5 cm)* failure progression


NSCLC and 13.3% IP 5pneumo-no
Lung progression drainage*
Metastases
20% EP Progression 4pneumonias*
(continued)
387
Table 1 (continued)
388

Author #/size LR Definition F/U (mn) Local Control Complications Survival


Patient
Pop.
Lee 2003 10/4.1 cm Any residual portion of lesion with 12.5 mn* 60% CR 10% major compl including mean
enhancement [ 10HU after 2 pneumo-thoracostomy; survival = 13.3 mn
contrast = viable tumor; 1ARDS*

NSCLC and Previously enhancing, but currently 40% PR


Lung unenhancing areas = necrosis;
Metastases
complete necrosis = nonenhancing Complete necrosis in all 6
area with a diameter [ initial viable tumors \ 3 cm, and in 23%(6/26)
tumor tumors [ 3 cm
Ambrogi 50(36 FU with contrast-enhanced ct scan, but 31* radiological CR-59%* 5/50-10% pneumothorax-2/ Median
2007 StageI)/ no definition of LR given 5 required pleural S = 25 mn*
NS, but drainage*
NSCLC all \ 5 cm

Different
Stages
Abbreviations
NSCLC = Non-small cell lung cancer
RECIST = Response evaluation criteria in solid tumors
CR = Complete response
CSS = Cancer specific survival
S = Survival
LR = Local recurrence
IP = Intrapulmonary recurrence
DR = Distal recurrence
DFS = Disease-free survival
Pneumo = Pneumothorax
PFS = Progression-free survival
EP = Extrapulmonary recurrence
* = Rates reported for total population of the study, not particularly for patients with Stage I NSCLC
LC = Local control
R = Recurrence
H&M nodes = Hilar and mediastinal nodes
NS = Not stated
AZ = Ablation zone
F/U = Follow-up
Mn = Months
J. M. Varlotto et al.
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer 389

Table 2 Summary of ablate and resect studies using RFA for treatment of lung neoplasms
Author Pt # with Tumor size Time Method of Definition Response Conclusion
population stage I interval assessment of response
NSCLC
Schneider* 14 1.7–3.5 immediate Histology-HE CN 5 CN CN in 35.7%
Schneider staining
et al. 2011

NSCLC and NADH Scattered 7 Scattered High rate of


Lung (Supravital vital tissue vital tissue tumor cells
Metastases staining)- remaining
performed in IA, [ 20% after RFA
22/32 vital tissue casts doubt
procedures 2 IA, [ 20% on RFA as
vital tissue curative
Monoclonal concept
anti-
mitochondrial IA did not
Antibodies- depend
performed in whether
18/32 primary or
procedures secondary, 2/
10 primary
lung and 2/7
mets with
adenoca had
IA
Ambrogi* 9 Max size of 5/9 Histology- Absence of CN in 6/9 Mean size
Ambrogi 3.5 cm immediate H&E staining tumor cells 2.3 cm with
et al. 2006 resection; with H&E CN

NSCLC- 6-Stage I Mean size 4/9


Stage I and II 3-StageII 2.8 cm resection Mean size of
after lesion with
15 days IA = 3.1 cm
All 1 cm
away from ALL IA-no
major blood more than
vessels and 10% of
airways treated area
with viable
tumor

Margin
\5 mm in all
three cases of
IA, average
margin 8 mm
in CA
Nguyen* 8 Mean tumor Immediate Histology- Absence of CA in 3-all Routine
Nguyen et al. size = 2.2 cm resection H&E cells by \2 cm; H&E
2005 NADH IA noted in 5 staining
staining tumors, 3 of could not
which were differentiate
\2 cm viable from
NSCLC- 7-StageI NADH nonviable
Stage I and II 1-Stage II cells
CN Complete necrosis, IA Incomplete ablation, CA Complete ablation
390 J. M. Varlotto et al.

treatment was to treat the tumor with a 5–10 mm margin metastatic lung tumors. The patients comprising the
beyond visible tumor in all directions (Beland et al. series by Ambrogi et al. (2006) and Nguyen et al.
2010; Pennathur et al. 2010). Most investigative series in (2005) were mostly patients with Stage I NSCLC.
Table 1 obtained CT scans immediately after treatment Complete ablation rates ranged from 35.7 to 66.7%.
to assess the extent of the GGA. The series (Ambrogi et al. 2006) with the highest rate
It is difficult to ascertain the benefit of RFA due to the of complete ablation used only hematoxylin and eosin
different radiologic definitions of local recurrence (LR) staining (H&E), a technique that others thought was
and because of the short follow-up of this population of unreliable (Nguyen et al. 2005; Schneider et al. 2011).
mostly poor-prognostic, medically-inoperable patients. The series of Schneider et al. (2011) used monoclonal
Additionally, two patient series treated selected patients anti-mitochondrial antibodies (MAM) and supravital
with adjuvant radiotherapy (Beland et al. 2010; Lanuti blue staining (SVB) in addition to routine H&E
et al. 2009). As shown in Table 1, median follow-up staining. Patients with complete necrosis were
ranged from 15 to 31 months. This limited follow-up required to have no evidence of tumor by either SVB
prevents adequate assessment of the effectiveness of or MAM, but it is not clear if the Stage I patients were
RFA as a method to obtain durable tumor control. assessed by MAM, SVB, or both. Although the
Although one series noted that the median time to local number of patients in each series is small, it appears
progression was only 9 months (Hiraki et al. 2007), that complete tumor necrosis was associated with size
another series noted a much longer median time to local in two studies (Ambrogi et al. 2006; Nguyen et al.
progression at 27 months (Pennathur et al. 2007). 2005) and ablation margins in one study (Ambrogi
Beland et al. (2010) noted that recurrences were spo- et al. 2006). Interestingly, Schneider et al. (2011)
radically identified throughout the 2 years of follow-up noted that all four tumors with incomplete ablation
and recommended continuous close follow-up during were adenocarcinomas (two metastatic colon cancer
this time. Size was noted to be related to LR, with sizes and two primary lung cancers). Complete necrosis
greater than 3 cm associated with LR in the series by and scattered vital tissue were noted in the other 15
Lee et al. (2003) and Lanuti et al. (2009). Another tumors of various histologies. Past pathologic reports
investigation noted that both size and stage were related have demonstrated that margins needed to encompass
to recurrence and, in addition, that there was non-sig- 95% of the primary NSCLC tumor extension were
nificant trend in favor of reduced recurrences in patients greater for adenocarcinoma (8 mm) than squamous
receiving external beam radiotherapy (RT) or brachy- cell carcinoma (6 mm) (Giraud et al. 2000). These
therapy (RT-br) (Beland et al. 2010). Nevertheless, results suggest that adenocarcinomas, whether pri-
local progression ranges from 20 to 42% at the local mary or metastatic, may require greater ablation
tumor site. Evaluation of treatment toxicity is rendered margins.
difficult due to the short patient follow-up, lack of Although the ablate and resect studies examined
assessment via an established toxicity scale, and the immediate tumor cell kill, RFA may be associated
retrospective nature of most series noted in Table 1. The with delayed tumor cell killing by stimulating the
most common complications include pneumothorax immune system. Therefore, assessment of tumor
(13–63%) and pleural effusion (10–21%). Pneumonias response by the pathologic assessment after immedi-
were reported in two of the series and occurred 16 and ate ablation may underestimate the responsiveness of
26.6% of the time, respectively. Other rare, but serious tumors to this technique. In one recent patient series
complications listed included hemorrhage, ARDS, and containing 14 primary and 6 metastatic lung tumors,
bronchopleural (bp) fistula. RFA led to an increase in neutrophils, monocytes, and
pro-inflammatory cytokines when serum was obtained
3 days after the procedure. Starting 30 days after
4.2 Ablate and Resect Studies RFA and persisting for up to 90 days after treatment,
further serum tests demonstrated a persistent reduc-
Results of the ablate and resect studies are listed in tion in the immunosuppressive CD 25 ? Foxp3
Table 2. Schneider et al. (2011) reported the results of T-regulatory cells with an increase in CD4 ?
32 patients, 14 of whom had Stage I non-small cell T-cell proliferation and the number of interferon
lung cancer (NSCLC) and the remaining patients had gamma-secreting cells (Fietta et al. 2009). Similarly,
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer 391

Fig. 4 a FDG-avid Stage I NSCLC presenting for RFA. b At 6 months there is complete loss of FDG avidity despite residual
mass on CT corresponding to complete tumor necrosis

RFA was associated with the elevation of tumor progressive contraction of the lesion and resolution of
antigen-specific antibodies in another patient series any pleural changes distal to the immediate ablation
(Widenmeyer et al. 2010). zone, as well as loss of contrast enhancement of the
lesion. A thin (\5 mm) pattern of peripheral/rim
enhancement, corresponding to reactive hyperemia, is
5 Follow-Up also associated with successful treatment (Anderson
et al. 2009). Cavities develop within the ablation zone
The appearances of treated lung cancer after RFA are in up to 20–58% of patients and are more commonly
well documented (Bojarski et al. 2005; Yasui et al. seen in lesions that significantly enlarge by more than
2004; Jin et al. 2004). The authors’ experience is to 200% from their original size (Fig. 6). Most patients
follow tumor size and contrast-enhancement patterns with cavities have no specific symptoms, and the
on CT Scans every 3 months for the first year and cavities usually spontaneously contract with time
twice a year thereafter. Loss of FDG-avidity at (Okuma et al. 2007; Steinke et al. 2003). Other
6 months on CT/PET can be used to confirm the reactive CT findings that are considered to be benign
absence of tumor when a residual mass is noted on CT include bubble lucencies and pleural thickening
scan (Fig. 4a–b). within or near the ablation zone (Pennathur et al.
Immediate CT following RFA demonstrates GGA 2007). When pleural effusions occur, they are usually
surrounding the tumor in 84% of cases and represents self-limiting, but can show FDG avidity (Higaki et al.
pulmonary bleeding and/or increased blood flow. 2008).
Within 1 week, the GGA changes to dense opacity The use of FDG-PET may prove to be a predictive
(Yasui et al. 2004). As noted previously, successful test for treatment failure or success. Higaki et al.
treatment is usually associated with a larger ablation (2008) suggest that immediate FDG PET with resid-
zone than the original tumor size (Yamamoto et al. ual activity before inflammation develops is likely to
2005) (Fig. 5). The ablation zone usually remains predict residual disease. Later assessment can dem-
larger during the first 3 months following the RFA onstrate three basic patterns as follows:
procedure. During this time period, transient and (1) A diffuse increase followed by continued
reversible locoregional nodal enlargement can some- decreasing activity.
times be appreciated (Sharma et al. 2010). After (2) Little change with sub-pleural lesions and rim
3 months, continued increase in growth of the abla- activity followed by a collapse of the lesion with
tion zone should be viewed as suspicious for incom- decreasing focal activity over time.
plete tumor destruction and recurrent tumor (Bojarski (3) Immediate residual activity or eccentric residual
et al. 2005). Successful treatment is associated with activity invariably showing progression over time.
392 J. M. Varlotto et al.

Fig. 5 Sequential CT scan following RFA of NSCLC. Site of tumor initially enlarges, followed by progressive contraction in
volume and changing shape over time. Note initial pleural thickening subsequently resolves
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer 393

Fig. 6 Small 7 mm squamous cell lung cancer. Note larger GGA with cavitary complication which progressively contracts

effusion, occurring in 13–63% and 10–21%, respec-


6 Summary tively. RFA is most effective with peripheral lesions
due to less concern for the heat-sink effect and due to
Since the first published report of modern percuta- the lower likelihood of complications. Prospective
neous RFA for the treatment of lung neoplasms in trials with careful long-term follow-up assessing diff-
2000, a number of largely retrospective studies have erent radiologic definitions of local control and using
demonstrated local control rates of 58–80%, with a known toxicity scale (i.e. common terminology
median follow-ups of 15–31 months. The primary criteria for adverse events (CTCAE v4.0)) are greatly
complications of RFA are pneumothorax and pleural needed.
394 J. M. Varlotto et al.

Due to the ability of SBRT to control greater 90% of for three-dimensional conformal radiotherapy planning. Int
primary tumors (Timmerman et al. 2010) and because J Radiat Oncol Biol Phys 48:1015–1024
Higaki T, Okumura Y, Sato S et al (2008) Preliminary
this technique does not require hospitalization, most retrospective investigation of FDG-PET/CT timing in
medically inoperable Stage I NSCLC patients are followup of ablated lung tumor. Ann of Nucl Med
treated with SBRT at Penn State Hershey Cancer 22:125–163
Institute. Central-based tumors within 2 cm of the Hiraki T, Gobara H, Iishi T et al (2007) Percutaneous
radiofrequency ablation for clinical stage I non-small cell
proximal bronchial tree (Timmerman et al. 2006) are lung cancer: results in 20 nonsurgical candidates. J Thorac
preferentially treated on RTOG 0813. RFA is consid- Cardiovasc Surg 134:1306–1312
ered for small (\1 cm) peripheral tumors in patients Jin GY, Lee JM, Lee YC, Han YM, Lim YS (2004) Primary
with limited prognosis due to the limitations of SBRT and secondary lung malignancies treated with percutaneous
radiofrequency ablation: evaluation with follow-up helical
including field dimensions of at least 3.5 cm (due to the CT. Am J Roentgenol 183:1013–1020
loss of electronic equilibrium with small beam aper- Koike T, Terashima M, Takizawa T, Watanabe T, Kurita Y,
tures) and possible greater incidence of rib pain. RFA Yokoyama A (1998) Clinical analysis of small-sized
is also considered for the rare patients experiencing peripheral lung cancer. J Thorac Cardiovasc Surg
115:1015–1019
primary tumor failure after SBRT. Lanuti M, Sharma A, Digumarthy SR et al (2009) Radiofre-
quency ablation for treatment of medically inoperable stage
I non-small cell lung cancer. J Thorac Cardiovasc Surg
References 137:160–166
Lee JM, Jin GY, Goldberg SN et al (2003) Percutaneous
radiofrequency ablation for inoperable non-small cell lung
Ambrogi MC, Fontanini G, Cioni R et al (2006) Biologic cancer and metastases: preliminary report. Radiology
effects of radiofrequency thermal ablation on non-small cell 230:125–134
lung cancer: results of a pilot study. J Thorac Cardiovasc Lencioni R, Crocetti L, Cioni R et al (2008) Response to
Surg 131:1002–1006 radiofrequency ablation of pulmonary tumors: a prospec-
Ambrogi MC, Dini P, Melfi F et al (2007) Radiofrequency tive, intention-to-treat, multicentre clinical trial (the rapture
ablation of inoperable non-small cell lung cancer. J Thorac study). Lancet Oncol 9:621–628
Oncol 2:S2–S3 Lilly MB, Brezovich IA, Atkinson W et al (1983) Hyperthermia
Anderson E, Lees W, Gillams A (2009) Early indicators of with implanted electrodes: in vitro and in vivo correlations.
treatment success after percutaneous radiofrequency of Int J Radiat Oncol Biol Phys 9:373–382
pulmonary tumors. Cardiovasc Intervent Radiol 32(3): Liu Z, Ahmed H, Iishi T et al (2006) Characterization of the RF
478–483 ablation-induced ‘‘oven effect’’: the importance of back-
Beland MD, Wasser EJ, Mayo-Smith WW et al (2010) Primary ground tissue thermal conductivity on tissue heating. Int J
non-small cell lung cancer: review of frequency, location, Hyperthermia 22:32–42
and time of recurrence after radiofrequency ablation. Matsuoka T, Okuma T (2007) CT-guided radiofrequency
Radiology 254:301–307 ablation for lung cancer. Int J Clin Oncol 12(2):71–78
Bojarski JD, Dupuy DE, Mayo-Smith WW (2005) CT imaging McTaggart RA, Dupuy DE (2007) Thermal ablation of lung
findings of pulmonary neoplasms after treatment with tumors. Tech Vasc Interv Radiol 10(2):102–113
radiofrequency ablation: results in 32 tumors. Am J Nguyen CL, Scott WJ, Young NA et al (2005) Radiofrequency
Roentgenol 185:466–471 ablation of primary lung cancer. Chest 128:3507–3511
Casal RF, Tam AL, Eapen GA (2010) Radiofrequency ablation Okuma T, Matsuoka T, Yamamoto A et al (2007) Factors
of lung tumors. Clin Chest Med 31:151–163 contributing to cavitation after CT guided percutaneous
Detterbeck FC, Jantz MA, Wallace M et al (2007) Invasive radiofrequency ablation of lung tumors. J Vasc Interv
mediastinal staging of lung cancer. Chest 132:202S–220S Radiol 18(3):399–404
Dupuy DE, Zagoria RJ, Akerley W et al (2000) Percutaneous Pennathur A, Luketich JD, Abbas G et al (2007) Radiofre-
radiofrequency ablation of malignancies in the lung. Am J quency ablation for the treatment of stage I non-small cell
Roentgenol 174:5–9 lung cancer in high-risk patients. J Thorac Cardiovasc Surg
Fietta AM, Morosini M, Passadore I et al (2009) Systemic 134(4):857–864
inflammatory response and down modulation of peripheral Pennathur A, Abbas G, Schuchert MJ et al (2010) Image-
CD25 ? Foxp3 ? T-regulatory cells in patients undergoing guided radiofrequency ablation for the treatment of early
radiofrequency thermal ablation for lung cancer. Hum stage non-small cell lung neoplasms in high-risk patients.
Immunol 70:47–86 Semin Thorac Cardiovasc Surg 22:53–58
Gilliams AR, Lees WR (2007) Analysis of the factors Rose SC (2008) Radiofrequency ablation of pulmonary
associated with radiofrequency ablation-induced pneu- malignancies. Semin Respir Crit Care Med 29:361–383
mothorax. Clin Radiol 62:639–644 Rose SC, Thistlethwaite PA, Sewell PE, Vance RB (2006)
Giraud P, Antoine M, Larrouy A et al (2000) Evaluation of Lung cancer and radiofrequency ablation. J Vasc Interv
microscopic tumor extension in non-small cell lung cancer Radiol 17:927–951
The Role of Radiofrequency Ablation in the Treatment of Stage 1 Non-Small Cell Lung Cancer 395

Rossi S, Dore R, Cascina A et al (2006) Percutaneous computed II study of stereotactic body radiation therapy for medically
tomography-guided radiofrequency thermal ablation of inoperable early-stage lung cancer. J Clin Oncol 24:
small unresectable lung tumors. Eur Respir J 27:556–563 4833–4839
Schneider T, Reuss D, Warth A et al (2011) The efficacy of Timmerman R, Paulus R, Galvin J et al (2010) Stereotactic
bipolar and multipolar radiofrequency ablation of lung body radiation therapy for inoperable early stage lung
neoplasms-results of an ablate and resect study. Eur J cancer. JAMA 303:1070–1076
Cardiothorac Surg in print White DC, D’Amico TA (2008) Radiofrequency ablation for
Sharma A, Digumarthy SR, Kalra MK et al (2010) Reversible primary lung cancer and pulmonary metastases. Clin Lung
locoregional lymph node enlargement after radiofrequency Cancer 9(1):16–23
ablation of lung tumors. Am J Roentgenol 194:1250–1256 Widenmeyer M, Shebzukhov Y, Haen SP et al (2010) Analysis
Steinke K, King J, Glenn D, Morris DL (2003) Radiologic of tumor antigen-specific T cells and antibodies in cancer
appearance and complications of percutaneous computed patients treated with radiofrequency ablation. Int J Cancer,
tomography-guided radiofrequency-ablated pulmonary in print
metastases from colorectal carcinoma. J Comput Assist Yamamoto A, Nakamura K, Matsuoka T et al (2005) Radio-
Tomogr 27:750–757 frequency ablation in a porcine lung model: correlation
Steinke K, Haghighi KS, Wulf S et al (2005) Effect of vessel between CT and histopathologic findings. Am J Roentgenol
diameter on the creation of ovine lung radiofrequency 185:1299–1306
lesions in vivo: preliminary results. J Surg Res 124:85–91 Yasui K, Kanazawa S, Sano Y et al (2004) Thoracic tumors
Timmerman R, McGarry R, Yiannoutsos C et al (2006) treated with CT-guided radiofrequency ablation: initial
Excessive toxicity when treating central tumors in a phase experience. Radiology 231:850–857
Lung Dose Escalation
Bradford S. Hoppe and Kenneth E. Rosenzweig

Contents Abstract
RTOG 73-01 established standard doses of radia-
1 Introduction.............................................................. 400 tion for the treatment of patients with stage III
2 Reducing Target Volumes ...................................... 400 non-small-cell lung cancer at 60 Gy in 2 Gy per
fraction. However, overall survival was still poor,
3 Reducing Normal-Tissue Irradiation .................... 401
and local failures were a continuing problem. Over
4 Early Studies ............................................................ 401 the next 30 years, a number of single institution
5 Radiation Dose Intensification Alone .................... 401 and multi institution studies have been performed,
6 Dose Escalation with Concurrent
attempting to improve overall survival by reducing
Chemotherapy .......................................................... 404 local failures through radiation dose escalation
either alone or in combination with chemotherapy
7 Current Status of Dose Escalation ........................ 407
with promising results. Additionally, new technol-
8 Conclusions ............................................................... 407 ogy has been developed that can improve tumor
References.......................................................................... 407 imaging, deliver more conformal RT with less
dose to normal structures, and decreased the set-up
uncertainties, which has increased the therapeutic
ratio and now allows for even safer dose escala-
tion. The present chapter reviews these studies and
discusses the current status of radiation dose
escalation for patients with stage III NSCLC.

Abbreviations

RTOG Radiation Therapy Oncology Group


NSCLC Non-small-cell lung cancer
RT Radiation therapy
B. S. Hoppe (&) Gy Gray
University of Florida Proton Therapy Institute,
SEER Surveillance, epidemiology and end
2015 North Jefferson St, Jacksonville,
FL 32206, USA result
e-mail: bhoppe@floridaproton.org CT Computed tomography
K. E. Rosenzweig FDG-PET Fluorodeoxyglucose positron emission
Mount Sinai School of Medicine, tomography
One Gustav L. Levy Place, Box 1236, GTV Gross tumor volume
New York, NY 10029, USA

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 399
DOI: 10.1007/174_2011_298, Ó Springer-Verlag Berlin Heidelberg 2011
400 B. S. Hoppe and K. E. Rosenzweig

ENI Elective nodal irradiation radiation delivery systems, and a better understanding
CTV Clinical tumor volume of the relationship between radiation dose-volume
PTV Planning target volume histograms of critical organs and treatment-related
kV Kilovoltage morbidity. As a result, safe dose escalation over
3D Three-dimensional 60 Gy is now more feasible (even with larger tumors)
4D Four-dimensional and a major focus of current clinical research studies.
PT Proton therapy
IMRT Intensity-modulated radiation therapy
BID Twice daily 2 Reducing Target Volumes
MTD Maximum tolerated dose
MSKCC Memorial Sloan–Kettering Cancer Target volumes have reduced over the last 20 years
Center due to reductions in: (1) gross tumor volume (GTV),
UM University of Michigan because of improved imaging, (2) clinical target
NKI Netherlands Cancer Institute volume (CTV), because of the elimination of elective
UNC University of North Carolina nodal irradiation, and (3) planning target volume
3D-CRT Three-dimensional conformal (PTV), because of reduced set-up uncertainty from
radiotherapy new imaging and immobilization techniques.
NTCP Normal-tissue complication probability Although computed tomography (CT) scanners
DVH Dose-volume histogram have been in use since 1973, it took close to 20 years
Veff V effective for them to become integrated into RT treatment
rMLD Relative mean lung dose planning. CT scans provide three-dimensional (3D)
CALGB Cancer and Leukemia Group B information regarding tumor volume as well as
NCCTG North Central Cancer Treatment Group demonstrate enlarged and pathologic lymph nodes.
An analysis based on surveillance, epidemiology and
end result (SEER) data showed that between 1994 and
2005, the use of CT-based simulation for the treat-
ment of thoracic malignancies increased from
1 Introduction 2.4 to 77.6%. Patients who had CT-based simula-
tion also had an improved survival as compared to
Poor outcomes in patients with unresectable non- patients who received conventional simulation
small-cell lung cancer (NSCLC) have been attrib- (Chen et al. 2011).
uted, in part, to low rates of local control with Over the last decade, fluorodeoxyglucose positron
traditional doses of definitive radiotherapy (RT). emission tomography (FDG-PET) imaging has also
Overall survival rates in NSCLC are expected to been used in conjunction with CT to aid in identifying
improve with better local control rates, which is malignant lymph nodes that were not enlarged
possible with higher radiation-dose levels. Unfortu- (negative on CT scan) and aid in separating the actual
nately, the therapeutic ratio for treating lung cancer tumor from atelectasis due to obstruction from the
with radiation has traditionally been small, due to tumor. Thus, GTVs have become easier to identify
the sensitivity of the lung and resulting pneumonitis and areas of uncertainties that were previously
and esophagitis with higher radiation doses. Thus, included in the GTV can now be more safely
dose escalation in patients with NSCLC has histor- eliminated.
ically been a double-edged sword with improved Historically, elective nodal regions have been
local control rates with higher doses, but corre- irradiated to doses of 45–50 Gy as part of the target
spondingly higher treatment-related morbidity and volume in patients with NSCLC. However, following
mortality. various series identifying distant and local relapses as
Over the last 20 years the therapeutic ratio of RT the primary sites of relapse, researchers began to
for NSCLC has widened due to reduced normal lung selectively eliminate the traditional elective nodal
tissue treated with high doses of radiation through a sites (Bradley et al. 2005; Rosenzweig et al. 2007;
reduction in the target volume, more sophisticated Hayman et al. 2001). Elimination of these elective
Lung Dose Escalation 401

nodal sites considerably reduces irradiation to normal set-up when the trial is under investigation. Most of
tissue, which can significantly impact treatment the current technologies used by treating physicians to
morbidity and mortality. In a phase III study from reduce both the dose to normal structures and the
China (Yuan et al. 2007), patients were randomized to margins of uncertainty in tumor delineation and set-
concurrent chemoradiation and either elective nodal up have only been included in the most recent studies
irradiation (ENI) with 60–64 Gy of RT or no ENI and that are now under investigation.
higher doses of RT (68–74 Gy). The group that
received ENI demonstrated higher rates of pneumo-
nitis (29 vs. 17%; p = 0.044), while better 2 year 4 Early Studies
overall survival rates were found in the group treated
with higher doses and no ENI (39.4 vs. 25.6%; Radiation Therapy Oncology Group (RTOG) trial
p = 0.048). As a result, most thoracic radiation 73-01 (Perez et al. 1980) was one of the first dose
oncologists are eliminating ENI from their treatment escalation studies. It not only established 60 Gy at
plan. 2 Gy per once-daily fraction as the standard of care
CTV and PTV margins in lung cancer treatment for unresectable NSCLC, it also pointed out glaring
have been reduced due to several innovations. Mar- problems in the management of NSCLC that required
gins for daily set-up errors have been minimized further investigation. The study randomized 376
through patient-specific immobilization devices, patients with T1-3 N0-2 NSCLC to either 40 Gy split
which, compared to no immobilization, are able to course (20 Gy at 4 Gy per fractions followed by a
more accurately reproduce patient positioning at the break followed by another 20 Gy at 4 Gy per frac-
time of simulation. Additionally, daily cone-beam CT tion), 40 Gy at 2 Gy per once-daily fraction, 50 Gy at
scans or orthogonal kV imaging allows for the repo- 2 Gy per once-daily fraction, or 60 Gy at 2 Gy per
sitioning of patients according to internal anatomy or once-daily fraction. Radiation techniques were two-
the actual tumor, thereby significantly reducing the dimensional (2D) and treatment fields included full
margins allotted for set-up errors compared with skin elective nodal irradiation. Intrathoracic relapses for
marks alone (Yeung et al. 2009). Lastly, with the use 40 Gy split, 40, 50, and 60 Gy were 44, 52, 42, and
of respiratory gating systems and 4D-CT scans, tumor 33%, respectively. Overall survival rates also
motion can be objectively measured and accounted improved with increasing radiation-dose levels at 24
for, which can lead to smaller margins. and 30 months. However, severe or life threatening
toxicity developed more frequently in the 60 Gy arm
with 14% compared with 5% of patients getting 50 Gy,
3 Reducing Normal-Tissue 7% of patients getting 40 Gy, and 13% in the 40 Gy
Irradiation split course. The study determined that 60 Gy should
be the standard radiation treatment dose with which
Three-dimensional treatment planning became pos- to compare future studies. Although higher doses were
sible when CT scans were integrated into radiation found to be associated with better outcomes, patients
treatment planning, allowing for more complex plans in both groups had high rates of extra-thoracic and
that better spared normal tissue and improved target unirradiated lung relapses. Thus, treatment intensifi-
coverage. Further improvements in target coverage cation was needed to improve outcomes.
and reductions in irradiated normal tissue have been
achieved with the implementation of more conformal
radiation delivery systems, including intensity- 5 Radiation Dose Intensification
modulated radiotherapy (IMRT), image guidance, and Alone
proton therapy (PT) (Sura et al. 2008; Bral et al. 2010;
Sejpal et al. 2011). These techniques are discussed in RTOG 83-11 (Cox et al. 1990) was a phase I/II
more detail in other chapters. hyperfractionation dose escalation study that ran-
Studies evaluating dose escalation in NSCLC dis- domized 884 patients with inoperable NSCLC to
cussed in this chapter must be considered in the either 60, 64.8, 69.6, 74.4, or 79.2 Gy at 1.2 Gy per
context of the limitations in technology and patient twice-daily (BID) fraction. The study took advantage
402 B. S. Hoppe and K. E. Rosenzweig

of the theoretical benefit of hyperfractionated radia- identifying local relapses in patients with lung cancer
tion to improve the therapeutic ratio, allowing for treated with RT.
safer dose escalation. Radiation was delivered to the Following this study, RTOG 93-11 evaluated dose
entire mediastinum, ipsilateral hilum, and contralat- escalation using once-daily doses of 2.15 Gy per
eral hilum to 50.4 Gy followed by a boost to the fraction (without inhomogeneity corrections) to find
primary lung cancer. Although rates of significant the maximum tolerated dose (MTD) (Bradley et al.
toxicities were similar amongst the different radiation 2005). The study allowed patients who had received
dose arms, overall survival rates for the entire cohort induction chemotherapy (*20%) and included 50%
were not significantly affected by radiation dose of patients with stage I disease and 50% with stage
either. In a subgroup of good performance status II–IIIB disease. This trial was based on previous data
patients, however, a survival advantage was seen at that correlated radiation pneumonitis with the volume
12 and 24 months with radiation doses of 69.6 Gy of lung receiving 20 Gy (Graham et al. 1999) and
or higher (p = 0.07), but there were no further placed 179 patients into one of three dose escalation
improvement in doses above 69.6 Gy. Comparing groups based on the lung V20. Patients with a lung
these results to RTOG 83-21, for which patients were V20 \ 25% received escalating doses to 70.9, 77.4,
randomized to standard radiation of 60 Gy at 2 Gy 83.8, and 90.3 Gy, while patients with a V20 between
per daily fraction with or without thymosin, a survival 25 and 36% received escalating doses to 70.9, 77.4,
advantage appeared likely with the hyperfractionated and 83.8 Gy. Furthermore, ENI was eliminated and
dose intensification of 69.6 Gy (median survival of all patients underwent CT-based treatment planning
8 months with 60 Gy at 2 Gy per daily fraction with the GTV delineating the tumor and enlarged
compared with 13 months with hyperfractionation). lymph nodes [1 cm and a 1 cm GTV to PTV margin
Based on the results of RTOG 83-11 and those expansion. The results from the study demonstrated
from the Cancer and Leukemia Group B (CALGB) that doses for patients with a V20 \ 25% could be
trial 84-33, which demonstrated a survival advantage safely escalated to 83.8 Gy with 15% rates of sig-
to induction chemotherapy prior to standard 60 Gy at nificant pulmonary toxicity at doses greater than
2 Gy per once-daily fraction (Dillman et al. 1996), the 77.4 Gy. In addition, those patients with a V20
RTOG developed protocol 88-08. RTOG 88-08 ran- between 25 and 36% could be escalated to 77.4 Gy
domized 498 patients with stage II–IIIB NSCLC with with 15% rates of pulmonary toxicity after doses
good performance status and \5% weight loss to greater than 70.4 Gy. Although dose did not affect
60 Gy at 2 Gy per once-daily fraction, 69.6 Gy at local–regional control or overall survival, the authors
1.2 Gy per BID fraction, or induction chemotherapy point out that those outcomes were not the main
of cisplatin and vinblastine followed by 60 Gy at objective of the study with only a few patients on
2 Gy per once-daily fraction (Sause et al. 1995). RT each of the many arms. Local–regional control still
was delivered with ENI to 50.4 Gy, followed by a remained a problem despite these higher doses of
boost to the primary disease and lymph nodes radiation: the overall rate of relapse was 38 and 18%
[2.5 cm in diameter as seen on pretreatment CT of patients experienced only a local failure. The
scan. The results showed a median survival of limited number of isolated regional failures (\10%)
11.4 months with 60 Gy of radiation alone and a confirmed that nodal relapses were rare and that ENI
marginal benefit with hyperfractionated radiation to could be excluded without negatively impacting dis-
69.6 Gy with a median survival of 12 months. ease control or overall survival. Due to the results
Importantly, induction chemotherapy followed by from this study and data emerging from other trials
60 Gy had a median survival of 13.2 months, thus evaluating concurrent chemotherapy and RT, the next
establishing a new standard of care for good perfor- stage of RTOG studies would involve radiation dose
mance patients with stage III NSCLC. Interestingly, escalation with concurrent chemotherapy.
when the pattern of relapses in this study was evalu- Memorial Sloan–Kettering Cancer Center
ated, the RTOG failed to identify any improvements (MSKCC; New York, NY), University of Michigan
in local control with induction chemotherapy, despite (UM; Ann Arbor), and The Netherlands Cancer
the improvement in overall survival (Komaki et al. Institute (NKI; Amsterdam) performed their own
1997). This finding elucidated the complexity of single institution dose escalation studies (Table 1).
Lung Dose Escalation 403

Table 1 Phase I dose escalation studies


Study/ RT ENI Chemotherapy Lung constraint # Of Lowest Highest MTD
Author technique patients dose (Gy) dose (Gy) (Gy)
UM 3D-CRT No Induction (*20%) Veff (0.32–0.4) 21 63 69 65.1
Veff (0.25–0.31) 18 63 75.6 75.6
Veff (0.19–0.24) 22 65.1 84 84
Veff (0.13–0.18) 20 69.3 102.9 102.9
Veff (0–0.12) 11 84 102.9 102.9
MSKCC 3D-CRT No Induction (16%) NTCP \ 25% 104 70.2 90 84
RTOG 3D-CRT No Induction (*15%) V20 \ 25% 127 70.9 90.3 83.8
9311
V20 (25–37%) 48 70.9 77.4 77.4
NKI 3D-CRT No Induction (18%) Veff (0.32–0.4) 3 60.75 60.75 60.75
Veff (0.25–0.31) 16 74.25 81 74.25
Veff (0.19–0.24) 23 74.25 87.75 81
Veff (0.13–0.18) 22 74.25 87.75 81
Veff (0–0.12) 24 81 94.5 94.5
China 3D-CRT Yes Induction and Adjuvant V20 \ 25% 15 72 78 78
V20 (25–37%) 20 72 78 78
V20 [ 37% 15 69 75 75
Carolinas 3D-CRT Yes Induction V35 \ FEV1*35 44 73.6 86.4 80
RTOG 3D-CRT No Concurrent V20 \ 30% 17 74 75.25 74
0117
UNC1 3D-CRT Yes Induction and V20 \ 35% 29 60 74 74
Concurrent
UNC2 3D-CRT Yes Induction and V20 \ 35% 29 78 90 90
Concurrent
NCCG 3D-CRT No Concurrent V20 \ 40% 15 70 78 74

From 1991 to 2003, MSKCC enrolled 104 patients, between 10 and 15 mm. Furthermore, the protocol
including 28% with stage I/II NSCLC and 68% with incorporated lung DVH data to ensure that the nor-
stage III NSCLC, on a three-dimensional conformal mal-tissue complication probabilities (NTCP) were
radiotherapy (3D-CRT) study (Rosenzweig et al. limited to \25%. The dose was escalated and the
2005). Patients were included even if they received MTD exceeded 90 Gy. Thus, 84 Gy was determined
induction chemotherapy (16%) and radiation was to be the MTD with only 1 of 21 patients developing a
delivered at 1.8 Gy once-daily fractions up to 81 Gy severe toxicity. Although this was a phase I study,
and then at 2 Gy once-daily fractions for higher doses dose was analyzed in relation to local control and
with a dose escalation scheme of 70.2, 75.6, 81, 84, overall survival. The investigators found that with
and 90 Gy (for smaller tumors only). The objective of doses of [80 Gy, overall survival was improved in
the study was to determine the MTD of 3D-CRT for patients with stage I/II disease (p = 0.05) and in
patients with NSCLC. ENI was included for the first patients with stage III disease (p = 0.02). Further-
20 patients treated to 70.2 Gy; however, three of these more, local control was improved with doses [80 Gy
patients developed severe toxicity (two grade 3 and (88%) compared with doses \80 Gy (14%).
one grade 5 pneumonitis), resulting in protocol UM’s phase I dose escalation study utilized lung
modifications. Subsequently, the target volume elim- DVH data to stratify patients into one of five separate
inated ENI and included the GTV to PTV margin dose escalation groups (Hayman et al. 2001; Narayan
404 B. S. Hoppe and K. E. Rosenzweig

et al. 2004). Dose escalation would begin at 84 Gy and CALGB 39904 was a prospective trial that asses-
end at 102.9 Gy with Veff \ 12% (bin 1), begin at sed accelerated, once-daily radiation therapy for early
69.3 Gy and end at 102.9 Gy with a Veff of 0.13–0.18 stage NSCLC (Bogart et al. 2010). The total dose of
(bin 2), begin at 65.1 Gy and end at 92.5 Gy with a Veff radiation remained at 70 Gy, but the dose per fraction
of 0.19–0.24 (bin 3), begin at 63 Gy and end at 92.4 Gy increased and the number of fractions decreased with
with a Veff of 0.25–0.31(bin 4), or begin at 63 Gy and each dose escalation level. The final cohort treated
end at 84 Gy with an NTCP[ 31% (bin 5). The target patients with 4.11 Gy per day for 17 fractions. The
volume included the gross tumor and any lymph nodes major response rate was 77% and the median survival
[1 cm on CT, which was then expanded to 0.5 cm was a favorable 38.5 months.
include the CTV. The PTV was created by expanding The Carolina Conformal Therapy Consortium per-
the CTV between 0.5 and 1 cm. The study enrolled 104 formed a phase I study from 1997 to 2002 of 44 patients
patients, including 25 who received induction chemo- with stage III NSCLC who were given induction che-
therapy (allowed after 1997). The study results deter- motherapy with carboplatin/paclitaxel or carboplatin/
mined an MTD of 65.1 Gy in patients in bin 5, while vinorelbine followed by accelerated hyperfractionated
they were 102.9, 102.9, 84, and 75.6 Gy for bins 1, 2, 3, 3D-CRT. Radiation targets included the
and 4, respectively. Interestingly, in those patients who GTV ? 0.5 cm for the CTV and another 0.5 cm for the
completed their prescribed radiation, the median sur- PTV. ENI was delivered at 1.25 Gy BID to a total dose
vival for stage I/II patients was 20 months and for stage of 45 Gy, while the PTV got 1.6 Gy BID to escalated
III patients it was 16 months. Although most relapses doses of 73.6, 80, and 86.4 Gy. The results demon-
occurred distantly (52%), 34% of the relapses did occur strated an MTD of 80 Gy, regardless of the chemo-
within the PTV alone. Eliminating the ENI did not therapy used. Despite ENI in this study, there were still
appear to affect outcomes given there were no isolated two regional failures in the 45 Gy region and five local
failures in the regional nodes, although three patients failures in the high-dose region.
relapsed in nodal regions simultaneously with local or Another study evaluating induction chemotherapy
distant failures. followed by radiation dose escalation came from
From 1998 to 2003, 88 patients were enrolled in a 3D- Fudan University (Shanghai, China). In this study, 50
CRT study by the NKI in a phase I/II dose escalation trial patients with stage II/III NSCLC were enrolled and
(Belderbos et al. 2006). Similar to the UM study, patients divided into one of three groups based on their V20
were separated into one of five risk groups based on their (\25, 25–37, and [37%). Mitomycin, vindesine, and
relative mean lung dose (rMLD). Induction chemo- cisplatin were given as induction chemotherapy and
therapy was allowed (18%) and the GTV was expanded then as adjuvant chemotherapy following RT. Dose
1–2 cm to create the PTV. No ENI was delivered. escalation started at 69 Gy in each arm and continued
3D-CRT was restricted to 6 weeks with a fixed frac- to 78 Gy at 3 Gy per fraction increments. The radi-
tion size of 2.25 Gy per fraction. If more than 30 ation volume included ENI to 42 Gy followed by a
fractions were needed, BID radiation was given. Group 1 boost to the GTV with a 1–1.5 cm margin. The MTD
(rMLD = 0–0.12) included only stage I (n = 19) or II was 78 Gy for the two groups with a lung V20 \ 37%
(n = 5) patients started at 81 Gy and was safely esca- and it was 75 Gy in the group with a V20 [ 37%.
lated to 94.5 Gy. Group 2 (rMLD = 0.12–0.18) and Unfortunately, despite this dose escalation with
Group 3 (rMLD = 0.18–0.24) started at 74.25 Gy and induction chemotherapy, the 2 year local–regional
went up to 87.75 Gy, but the MTD was reached at progression-free survival was 41%, leaving room for
81 Gy. Group 4 (rMLD = 0.24–0.31) started at improvement.
74.25 Gy and was escalated to 81 Gy, but the MTD was
reached at 74.25. Group 5 (rMLD = 0.31–0.4) only
enrolled three patients and was safely treated to 6 Dose Escalation with Concurrent
60.75 Gy. Although dose could safely be escalated using Chemotherapy
3D-CRT, no advantage to a higher dose was found in any
group aside from Group 1. In this study, eliminating ENI A number of studies have evaluated concurrent che-
resulted in few nodal failures and the local–regional moradiation for locally advanced NSCLC. The RTOG
failure rate was 28%. began its chemoradiation studies with RTOG 88-04,
Lung Dose Escalation 405

which was a phase II study evaluating induction The local–regional control is thought to have not
cisplatin and vinblastine followed by concurrent translated into improved overall survival rates because
cisplatin with once-daily RT to 60 Gy. This was of the increased 68% grade 3+ nonhematologic toxicity
followed up by RTOG 90-15 with concurrent cisplatin experienced in the RT dose intensification arm com-
and vinblastine with dose escalation using hyper- pared with 48% in arm 2. The investigators thus con-
fractionated RT to 69.6 Gy. Due to the excessive rate cluded that with crude 2D-RT techniques and large
of hematologic toxicity in that study, it was subse- treatment volumes, RT dose intensification above
quently followed up by RTOG 91-06, a study evalu- 60 Gy was not possible. Other investigators have
ating a less aggressive chemotherapy, concurrent confirmed the improvement in overall survival dem-
cisplatin with daily etoposide with BID RT to onstrated with concurrent chemoradiation.
69.6 Gy. Due to the promising results from 88-04 and Although RTOG 94-13 failed to demonstrate an
91-06, a phase II randomized trial, RTOG 92-04, was improvement in survival with concurrent chemother-
performed comparing these two regimens (Komaki apy and dose escalation to 69.6 Gy BID compared with
et al. 2002). This study demonstrated reduced in-field 63 Gy given once a day, data emerging from RTOG
relapse rates with the RT dose escalation to 69.6 Gy 93-11 demonstrated the ability to safely dose escalate
BID, but also a significantly increased risk of early through the use of 3D-CRT and the elimination of ENI.
and late esophagitis with this treatment arm. Thus, RTOG 0117 was developed to evaluate dose
RTOG 94-10 randomized 595 patients with stage escalation with concurrent chemotherapy utilizing
II–III NSCLC to the newly established gold standard: modern RT techniques. (Table 2) In the phase I portion
induction chemotherapy followed by 60 Gy at 2 Gy per of the study, 17 patients with stage I–IIIB NSCLC
fraction (arm 1; similar to CALGB 84-33 and RTOG were enrolled with treatment consisting of weekly
88-08), to concurrent chemotherapy and 60 Gy at 2 Gy paclitaxel 50 mg/m2 and carboplatin area under the
per fraction (arm 2), and to concurrent chemotherapy concentration (AUC) of two given concurrently with
with RT dose escalation to 69.6 Gy at 1.2 Gy per BID escalating doses of RT. Initially, patients were to
fraction (arm 3). Chemotherapy consisted of cisplatin receive 75.25 Gy at 2.15 Gy per fraction, 80.5 Gy at
100 mg/m2 and vinblastine 5 mg/m2 on days 1 and 29 2.3 Gy per fraction, 79.5 at 2.65 Gy per fraction, and
in the daily RT arms, while the third arm of BID RT 75 Gy at 3 Gy per fraction; however, because of
received cisplatin 50 mg/m2 on days 1, 8, 29, and 36, excessive toxicity at the first dose level, the dose
and oral etoposide to 50 mg given BID for 10 weeks. changed to 74 Gy at 2 Gy per fraction. All patients
RT was delivered using 2D techniques to large treat- received 3D-CRT and the target volume was similar to
ment volumes including the elective nodal regions. The RTOG 93-13. Patients were also required to have a lung
primary objective of the trial was to evaluate whether V20 \ 30%, mean esophagus dose \34 Gy, and an
concurrent chemotherapy and RT had a better overall esophageal V55 of \30%. Due to excessive toxicity in
survival than induction chemotherapy followed by RT. the 75.25 Gy arm, the study was de-escalated to 74 Gy,
Additionally, it was evaluating whether RT dose at which acceptable toxicity occurred with only one
escalation to 69.6 Gy given BID concurrently with grade 4 event in seven patients (Bradley et al. 2010).
chemotherapy would be better than concurrent che- The phase II portion of RTOG 0117 borrowed the
moradiation to 60 Gy. Although the results have never 74 Gy arm at 2 Gy per fraction from the phase I portion
been formally published as a manuscript, data reported (n = 9) and enrolled another 46 patients. The results
in an abstract, demonstrated median survivals of 14.6, demonstrated a median survival of 21.6 months for
17, and 15.2 months for arms 1, 2, and 3, respectively patients with stage III NSCLC, which compared quite
(Curran 2000). Although the study confirmed that favorably to the concurrent chemoradiation arm to
concurrent chemoradiation was better than induction 63 Gy from RTOG 9410 (17 months). Furthermore,
chemotherapy followed by RT, it did not demonstrate only 12 patients experienced grade 3 or higher lung
improvement with RT dose intensification to 69.6 Gy toxicity and 21 patients experienced grade 2 or higher
BID over 60 Gy daily in overall survival. However, esophagitis (Bradley et al. 2010).
it did demonstrate improved local–regional control While RTOG 0117 included dose escalation with
with the more intensified RT: 34% compared with concurrent chemotherapy, the North Central Cancer
43% in the concurrent chemotherapy arm to 60 Gy. Treatment Group (NCCTG) and the University of
406 B. S. Hoppe and K. E. Rosenzweig

Table 2 RTOG non-small cell lung cancer studies


Study/ Patients RT dose RT ENI Chemotherapy Median Acute (%) Late (%)
Author technique OS Grade 3/4/5 toxicity
non-hematologic
RTOG 7301 97 40 Gy split 2D Yes None 9.3 13
103 40 Gy 10 7
91 50 Gy 11 6
85 60 Gy 12 14
RTOG 83-11 83 60 Gy BID 2D Yes None 9.2 7.0 8.6
127 64.8 Gy BID 6.3 13.0 5.7
220 69.6 Gy BID 10 10.0 8.5
211 74.4 Gy BID 8.7 12.0 7.0
207 79.2 Gy BID 10.5 11.0 10.7
RTOG 88-08 60 Gy 2D Yes None 11.4 NA NA
60 Gy Yes Induction 13.8 NA NA
69.6 Gy BID Yes None 12.3 NA NA
RTOG 9204 81 63 Gy 2D Yes Induction and 16.4 35.0 25.0
concurrent
81 69.6 Gy BID 2D Yes Concurrent 15.5 54.0 40.0
RTOG 9410 201 60 Gy 2D Yes Induction 14.6 NA NA
201 60 Gy Concurrent 17 48.0 NA
193 69.6 Gy BID Concurrent 15.2 68.0 NA
RTOG 0117 55 74 Gy 3D No Concurrent 21.6 61.0 22.0

North Carolina (UNC; Chapel Hill) conducted their the phase I arm of the study. On this arm, dose escalation
own protocols investigating dose escalation with began at 60 Gy and was followed by doses of 66, 70, and
concurrent chemotherapy. The NCCTG 0028 was a 74 Gy. Once 74 Gy had been reached and was deter-
phase I study where 15 patients with unresectable mined to be safe (there was only one case of grade 3
NSCLC underwent dose escalation from 70 to 78 Gy esophagitis), the phase II component began, with the
in 4 Gy increments. RT was delivered using 3D-CRT, enrollment of an additional 33 patients to receive 74 Gy.
limiting the lung V20 \ 40%, and no ENI was The median survival was 24 months with only 8 patients
delivered. The results found the MTD to be 74 Gy in (13%) developing local–regional relapse as the only
combination with weekly carboplatin (AUC 2) and site of failure, although 35% of patients had a local
pacilitaxel (50 mg/m2), which were consistent with failure as a component of their relapse. RTOG grade 3/
the findings of RTOG 0117. 4 esophgagitis developed in only five patients (8%) and
Between 1996 and 1999, 62 patients were enrolled no grade 3/4 pneumonitis developed.
on a phase I/II study conducted by UNC that Considering the promising results of their dose
included stage III NSCLC (Rosenman et al. 2002). escalation study, UNC developed a follow-up study
Patients received induction chemotherapy (carboplatin/ that escalated the radiation dose from 78 Gy to 82, 86,
paclitaxel) followed by concurrent weekly carboplatin/ and 90 Gy (Fried et al. 2004). Twenty-nine patients
paclitaxel with escalating 3D-CRT doses from 60 to were enrolled in this study and all patients received
74 Gy. ENI was performed to doses between 46 and induction chemotherapy consisting of carboplatin
50 Gy. A generous margin of at least 1.5 cm was gen- (AUC = 5), irinotecan (100 mg/m2), and paclitaxel
erated to create the PTV. The first 29 patients comprised (175 mg/m2) followed by concurrent chemotherapy
Lung Dose Escalation 407

(carboplatin AUC 2 and paclitaxel 45 mg/m2 weekly). this study should guide researchers towards the next
Although 3D-CRT was used for radiation treatment step in phase I and II studies for NSCLC.
planning, the study also included ENI to doses between
40 and 50 Gy. The study safely escalated the dose from
78 to 90 Gy without reaching the MTD. Acute grade 3
esophagitis developed in 16% of patients. Late toxici- References
ties consisted of one patient with grade 3 pneumonitis
that resolved and two patients with fatal hemoptysis. Belderbos JS, Heemsbergen WD, de Jaeger K, Baas P,
Lebesque JV (2006) Final results of a Phase I/II dose
escalation trial in non-small-cell lung cancer using three-
dimensional conformal radiotherapy. Int J Radiat Oncol
7 Current Status of Dose Escalation Biol Phys 66:126–134
Bogart JA et al (2010) Phase I study of accelerated conformal
Due to the successes with RTOG 0117, NCCTG0024, radiotherapy for stage I non-small-cell lung cancer in
patients with pulmonary dysfunction: CALGB 39904. J Clin
and the UNC dose escalation studies, RTOG currently
Oncol: Off J Am Soc Clin Oncol 28(2):202–206
has a randomized controlled trial, RTOG 0617, to Bradley J, Graham MV, Winter K, Purdy JA, Komaki R, Roa
confirm the benefits of radiation dose escalation with WH, Ryu JK, Bosch W, Emami B (2005) Toxicity and
concurrent chemotherapy. The study, which is cur- outcome results of RTOG 9311: a phase I–II dose escalation
study using three-dimensional conformal radiotherapy in
rently ongoing, is a 2 9 2 study on which patients
patients with inoperable non-small-cell lung carcinoma. Int
with stage III NSCLC are randomized to between 60 J Radiat Oncol Biol Phys 61:318–328
and 74 Gy of radiation with concurrent weekly pac- Bradley JD, Moughan J, Graham MV, Byhardt R, Govindan R,
litaxel and carboplatin with or without cetuximab. Fowler J, Purdy JA, Michalski JM, Gore E, Choy H (2010)
The study’s main objective is to evaluate for A phase I/II radiation dose escalation study with concurrent
chemotherapy for patients with inoperable stages I to III
improved overall survival and is aimed at enrolling non-small-cell lung cancer: phase I results of RTOG 0117.
500 patients. Radiation is being given on the study Int J Radiat Oncol Biol Phys 77:367–372
either by 3D-CRT or IMRT and ENI is not being Bral S, Duchateau M, Versmessen H, Verdries D, Engels B, De
delivered. Patients on this study will be undergoing Ridder M, Tournel K, Collen C, Everaert H, Schallier D, De
Greve J, Storme G (2010) Toxicity report of a phase 1/2
PET-CT scan as part of their staging. An important dose escalation study in patients with inoperable, locally
aspect of this study is its use of some of the most up- advanced nonsmall cell lung cancer with helical tomother-
to-date radiation delivery systems (i.e., IMRT) and apy and concurrent chemotherapy. Cancer 116:241–250
imaging scans (i.e., PET-CT scan) available. Thus, Chen AB et al (2011) Survival outcomes after radiation therapy
for stage iii non-small-cell lung cancer after adoption of
the study results will be highly relevant to the future computed tomography-based simulation. J Clin Oncol: Off j
direction of cooperative groups. Am Soc Clin Oncol 40:464
Cox JD, Azarnia N, Byhardt RW, Shin KH, Emami B, Pajak TF
(1990) A randomized phase I/II trial of hyperfractionated
radiation therapy with total doses of 60.0–79.2 Gy: possible
8 Conclusions survival benefit with greater than or equal to 69.6 Gy in
favorable patients with radiation therapy oncology group
Safer dose escalation with RT beyond standard doses of stage III non-small-cell lung carcinoma: report of Radiation
radiation established by RTOG 73-01 has been possible Therapy Oncology Group 83-11. J Clin Oncol 8:1543–1555
Curran W (2000) Phase III comparison of sequential vs.
with advances in imaging and radiation technology. concurrent chemoradiation for patients with unresected
Higher doses of radiation, even with the omission of stage III non-small-cell lung cancer (NSCLC): initial report
ENI, appear to improve local–regional control; how- of radiation therapy oncology group (RTOG) 9410. Pro Am
ever, overall survival and distant metastases remain a Soc Clin Oncol 19S:1891a
Dillman RO, Herndon J, Seagren SL, Eaton WL Jr, Green MR
huge problem. Although dose escalation with concur- (1996) Improved survival in stage III non-small-cell lung
rent chemotherapy has been possible, outcomes from cancer: seven-year follow-up of cancer and leukemia group
phase I and II studies have not been confirmed in a B (CALGB) 8433 trial. J Natl Cancer Inst 88:1210–1215
phase III study. The current RTOG 0617 study is Fried DB, Morris DE, Poole C, Rosenman JG, Halle JS,
Detterbeck FC, Hensing TA, Socinski MA (2004) System-
addressing these critical issues in a randomized con- atic review evaluating the timing of thoracic radiation
trolled trial while also investigating one of the newest therapy in combined modality therapy for limited-stage
targeted agents in lung cancer: cetuximab. Results from small-cell lung cancer. J Clin Oncol 22:4837–4845
408 B. S. Hoppe and K. E. Rosenzweig

Graham MV, Purdy JA, Emami B, Harms W, Bosch W, results of a phase I/II trial. Int J Radiat Oncol Biol Phys
Lockett MA, Perez CA (1999) Clinical dose-volume 54:348–356
histogram analysis for pneumonitis after 3D treatment for Rosenzweig KE, Fox JL, Yorke E, Amols H, Jackson A,
non-small cell lung cancer (NSCLC). Int J Radiat Oncol Rusch V, Kris MG, Ling CC, Leibel SA (2005) Results of a
Biol Phys 45:323–329 phase I dose escalation study using three-dimensional
Hayman JA, Martel MK, Ten Haken RK, Normolle DP, conformal radiotherapy in the treatment of inoperable
Todd RF, Littles JF, Sullivan MA, Possert PW, Turrisi AT, nonsmall cell lung carcinoma. Cancer 103:2118–2127
Lichter AS (2001) Dose escalation in non-small-cell lung Rosenzweig KE, Sura S, Jackson A, Yorke E (2007) Involved-
cancer using three-dimensional conformal radiation ther- field radiation therapy for inoperable non small-cell lung
apy: update of a phase I trial. J Clin Oncol 19:127–136 cancer. J Clin Oncol 25:5557–5561
Komaki R, Scott CB, Sause WT, Johnson DH, Taylor SG, Sause WT, Scott C, Taylor S, Johnson D, Livingston R,
Lee JS, Emami B, Byhardt RW, Curran WJ Jr, Dar AR, Komaki R, Emami B, Curran WJ, Byhardt RW, Turrisi AT
Cox JD (1997) Induction cisplatin/vinblastine, irradiation et al (1995) Radiation therapy oncology group (RTOG)
vs. irradiation in unresectable squamous cell lung cancer: 88–08 and eastern cooperative oncology group (ECOG)
failure patterns by cell type in RTOG 88–08/ECOG 4588. 4588: preliminary results of a phase III trial in regionally
Radiation Therapy Oncology Group: Eastern Cooperative advanced, unresectable non-small-cell lung cancer. J Natl
Oncology Group. Int J Radiat Oncol Biol Phys 39:537–544 Cancer Inst 87:198–205
Komaki R, Seiferheld W, Ettinger D, Lee JS, Movsas B, Sejpal S, Komaki R, Tsao A, Chang JY, Liao Z, Wei X,
Sause W (2002) Randomized phase II chemotherapy and Allen PK, Lu C, Gillin M, Cox JD (2011) Early findings on
radiotherapy trial for patients with locally advanced inop- toxicity of proton beam therapy with concurrent chemo-
erable non-small-cell lung cancer: long-term follow-up of therapy for nonsmall cell lung cancer. Cancer
RTOG 92-04. Int J Radiat Oncol Biol Phys 53:548–557 Sura S, Gupta V, Yorke E, Jackson A, Amols H, Rosenzweig KE
Narayan S, Henning GT, Ten Haken RK, Sullivan MA, (2008) Intensity-modulated radiation therapy (IMRT) for
Martel MK, Hayman JA (2004) Results following treatment inoperable non-small cell lung cancer: the Memorial Sloan-
to doses of 92.4 or 102.9 Gy on a phase I dose escalation Kettering Cancer Center (MSKCC) experience. Radiother
study for non-small cell lung cancer. Lung Cancer 44:79–88 Oncol 87:17–23
Perez CA, Stanley K, Rubin P, Kramer S, Brady L, Perez- Yeung AR, Li JG, Shi W, Newlin HE, Chvetsov A, Liu C,
Tamayo R, Brown GS, Concannon J, Rotman M, Seydel HG Palta JR, Olivier K (2009) Tumor localization using cone-
(1980) A prospective randomized study of various irradiation beam CT reduces setup margins in conventionally fraction-
doses and fractionation schedules in the treatment of ated radiotherapy for lung tumors. Int J Radiat Oncol Biol
inoperable non-oat-cell carcinoma of the lung: preliminary Phys 74:1100–1107
report by the Radiation Therapy Oncology Group. Cancer Yuan S, Sun X, Li M, Yu J, Ren R, Yu Y, Li J, Liu X, Wang R,
45:2744–2753 Li B, Kong L, Yin Y (2007) A randomized study of
Rosenman JG, Halle JS, Socinski MA, Deschesne K, involved-field irradiation versus elective nodal irradiation in
Moore DT, Johnson H, Fraser R, Morris DE (2002) combination with concurrent chemotherapy for inoperable
High-dose conformal radiotherapy for treatment of stage stage III non-small-cell lung cancer. Am J Clin Oncol
IIIA/IIIB non-small-cell lung cancer: technical issues and 30:239–244
Radiochemotherapy in Locally Advanced
Non-small-Cell Lung Cancer
Branislav Jeremić, Francesc Casas, and Asuncion Hervas-Moron

Contents Abstract
Locally advanced nonsmall cell lung cancer is one
1 Introduction.............................................................. 409 of the major battlegrounds in clinical research in
2 Radiation Therapy Alone ....................................... 410 lung cancer due to opportunities for all the treatment
options to be employed either alone or in various
3 Neoadjuvant (Induction) Chemotherapy
Followed by Radiation Therapy ............................ 412 combinations in both curative and palliative setting.
In curative setting, though, recent evidence recon-
4 Concurrent Radiochemotherapy............................ 415
firms advantage of concurrent radiochemotherapy
5 Neoadjuvant (Induction) Chemotherapy over other existing treatment options. It is expected
Followed by Radiation Therapy Versus that with novel radiotherapy technologies and new
Concurrent Radiochemotherapy............................ 417
generations of drugs overall results in this disease
6 Optimization of Concurrent are further improved.
Radiochemotherapy ................................................. 420
7 New Approaches in Radiation Therapy
and Chemotherapy of Locally Advanced
Nonsmall-Cell Lung Cancer................................... 423
1 Introduction
8 Conclusions ............................................................... 425
References.......................................................................... 425 Approximately one-third of all patients with nonsmall
cell lung cancer present with a locally advanced,
mostly stage III disease. Patients falling into this
group represent a heterogeneous group of patients.
According to the two staging systems widely adopted
in the community of thoracic oncologists (Mountain
1986, 1997), this is almost synonymous to stage III,
although numerous, mostly non-surgical reports,
included also a proportion of patients with stage II
disease into this group of patients. The most recent
attempt of the International Association for the Study
of Lung Cancer included also non-US and non-sur-
B. Jeremić (&)
Institute of Lung Diseases, Sremska Kamenica, Serbia gical patients in order to increase the overall number
e-mail: nebareje@gmail.com of patients suitable to analysis, increase statistical
F. Casas power to detect small difference, improve sensitivity
University Clinic, Barcelona, Spain analysis and, hence, provide necessary evolution
A. Hervas-Moron of the staging principles in lung cancer. The series of
Hospital Ramon y Cajal, Madrid, Spain articles extensively documented various aspects of

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 409
DOI: 10.1007/174_2011_232, Ó Springer-Verlag Berlin Heidelberg 2011
410 B. Jeremić et al.

this important issue (Goldstraw and Crowley 2006; disease. This, however, is not so frequent practice
Groome et al. 2007; Rami-Porta et al. 2007; Rusch nowadays in the majority of patients who can tolerate
et al. 2007; Postmus et al. 2007; Goldstraw et al. more intensive (combined) treatment approach owing
2007). It should be noted that both previous and to the mounting evidence that they could successfully
current staging systems were nevertheless surgical, i.e. be treated with such a combined modality approach.
those in which major principle remained the ability to In practice this means, a bimodality (radiochemo-
surgically extirpate of the tumour and lymph nodes. therapy) or a trimodality approach, the latter one
The only issues which do matter in these systems is the being detailed in another chapter.
size of the tumour (e.g. 3 or 5 cm in largest diameter),
its location (e.g. 2 cm or more from carina), and inva-
sion of neighbouring structures (e.g. chest wall or 2 Radiation Therapy Alone
heart). These systems even nowadays, unfortunately,
do not take into account tumour volumes at all! That Locally advanced nonsmall cell lung cancer has fre-
said, a major principle of anticancer action of both quently been treated with radiation therapy alone. Due
radiation therapy and chemotherapy, and that is log-cell to its characteristics, radiation therapy was relatively
kill, is not considered at all. This is an extremely well tolerated. It offered good palliation and since there
important issue, especially in a number of tumours have been very few alternatives, only a few studies
which could have similar tumour volume, but which validating its efficacy exist. More than 40 years ago, the
could have been designed a different stage in case of Veterans’ Affairs group reported on a study where
different location or invasion of neighbouring struc- radiation therapy had been compared to best supportive
tures. This is especially important in stage III nonsmall care for locally advanced nonsmall cell lung cancer.
cell lung cancer which has also underwent a change in Radiation therapy led to an improvement in the median
the most recent staging revision. It still, however, has survival time, but no 2-year survivors were seen in
11 (!) different T and N combinations, ranging from either arm (Roswit et al. 1968). Legitimate criticism of
T4N0 to T1N3. The contribution of T and N compo- this study include rather primitive staging procedures
nent, to a particular stage obviously greatly vary. (many patients in both arms would likely have Stage IV
Although stage subgroupings should reflect similar disease if modern imaging was used), and outdated
prognosis, one crucial issue remained unanswered: is radiation therapy techniques (from today’s standpoint).
there a ‘‘effect trade-off’’ when adverse effect of an More recently, however, similar results were obtained
increasing T stage is, perhaps, levelled-off with in another randomized study. The 2-year survival was
decreasing N stage, or perhaps which one of these two 18% with radiation therapy dose of 50 Gy versus 0%
descriptors may be more important than the other one for observation with palliative radiation therapy
and if so, then exactly when? Finally, since volumes of administered when severe local symptoms developed
these descriptors could also vary (depending on their (Reinfuss et al. 1999). In another randomized cooper-
size, and/or location), the nature of non-surgical cell ative group study from the US patients were random-
kill (i.e. log cell kill) favours the volume use in this ized to radiation therapy versus single agent vindesine
discussion. It is expected that future staging system (Johnson et al. 1990), while the third arm contained
revisions may start taking into account tumour volume, both modalities. The study showed no survival advan-
as should be much easier to do nowadays due to pow- tage for radiation therapy. However, in this study there
erful computers used for CT, MRI and PET scanning. was a substantial crossover in this trial, with many
Locally advanced nonsmall cell lung cancer has patients in the vindesine arm ultimately receiving
been the major battleground for investigating various thoracic radiation therapy. Although this study sug-
treatment options. Surgery (e.g. in very selected gested no survival advantage to ‘‘early’’ radiation
T4N0), radiation therapy (altered fractionation regi- therapy, this should not imply no advantage at all for
mens with curative intention in stage III or palliative radiation therapy.
hypofractionated regimens in mostly stage IIIB Regardless of the shortcomings from the studies,
patients) and chemotherapy (in stage IIIB in numer- radiation therapy has been considered as the mainstay
ous clinical studies investigating effect of various of therapy for locally advanced non-small cell lung
chemotherapeutic agents) can all be used alone in this cancer in the past. In the benchmark trial, continuous
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 411

course of radiation therapy with doses of 50–60 Gy as hyperfractionated accelerated radiation therapy
has been shown to be superior to either 40 Gy split- (Mehta et al. 1998) which also proven to be effective
course or continuous course schedule (Perez et al. in this setting.
1986). The 60 Gy-continuous course schedule has Studies using altered fractionated regimens
then been adopted as the standard radiation therapy all reconfirmed the duality of patterns of failure in
over the world. However, the results obtained with patients treated with radiation therapy alone. These
radiation therapy alone given that way were unsatis- facts again stressed the need for inclusion of chemo-
factory, since the median survival time was approxi- therapy into the overall treatment plan: to improve
mately 9–10 months and the overall 5-year survival both local/regional control and combat possible dis-
rate was only 3–6% in prospective randomized trials tant (microscopic) spread, the latter not addressed by
(Holsti and Matson 1980; Petrovich et al. 1981; Perez the radiation therapy. Unfortunately, the results of
et al. 1987). Since various retrospective and pro- early studies designed with that aim were neither
spective randomized studies have revealed that both encouraging nor different from that obtained with
local and systemic failure play an important role in radiation therapy alone. Timing of chemotherapy
the poor survival of these patients (Petrovich et al. administration was usually adjuvant (i.e. post-radia-
1977; Cox et al. 1979; Perez et al. 1986), various tion therapy) and it consisted of non-platinum based
means of improving local and systemic control of this drugs (Reynolds and O’Dell 1978; White et al. 1982).
disease were sought. Radiation therapy-induced fibrotic changes in lungs
In the domain of radiation therapy alone, altered that prevented successful blood/drug perfusion and,
fractionation regimens have been used to improve therefore, drug supply to the tumour-bearing area,
local control (Cox et al. 1993; Saunders and Dische was the main reason for such observation. Relative
1990; Byhardt et al. 1993). The Radiation Therapy inefficiency of then-available drugs, mostly consid-
Oncology Group (Cox et al. 1990) has investigated ered as first-generation chemotherapy drugs, origi-
hyperfractionated radiation therapy with 1.2 Gy b.i.d. nating in the pre-cisplatin era (Reynolds and O’Dell
fractions and reported improved survival in a sub- 1978; White et al. 1982) was also implicated in this
group of patients with favourable prognostic factors inefficiency.
treated with hyperfractionated radiation therapy with Contrary to this, radiation therapy and platinum-
doses C69.6 Gy compared to that obtained with the based chemotherapy have been increasingly practiced
standard treatment (60 Gy/30 fractions/6 weeks). around the world in the last three decades. A number
Continuous hyperfractionated accelerated radiation of possible combinations have arisen, largely
therapy (CHART) was tested against standard frac- exploiting different aspects of such combination,
tionation radiation therapy in inoperable non-small- frequently focusing on the issue of timing/scheduling.
cell lung cancer and it was shown be beneficial Induction (neo-adjuvant) chemotherapy followed by
(Saunders et al. 1999). This treatment design was, radical radiation therapy (Dillman et al. 1990; Sause
unfortunately, extremely complicated for daily clini- et al. 1995), ‘‘sandwich’’ chemotherapy and radiation
cal practice which has prevented it from widespread therapy (Le Chevalier et al. 1992) as well as con-
use, even in the UK. Several attempts to modify it current radiochemotherapy (Schaake-Koning et al.
included the omission of week-end days or neoadju- 1992; Jeremic et al. 1995, 1996, 1998) have all gained
vant chemotherapy, both of which effectively destroy widespread use, sometimes with very similar results
its underlying principle and that was accelerated obtained with these approaches, with quite different
fractionated radiation therapy to combat accelerated radiobiological background. To further obscure the
tumour clonogen proliferation. It was then not sur- overall picture, both radiation therapy and chemo-
prising that the CHART Weekend-less CHARTWEL therapy have evolved over the years. A number of
trial which compared 66 Gy in 33 daily fractions with different time/dose/fractionation radiation therapy
60 Gy in 40 fractions in 18 treatment days (t.i.d.) regimens have been used (Cox et al. 1990; Byhardt
has not found a survival advantage for accelerated et al. 1993; Saunders and Dische 1990). They paral-
regimen (Baumann et al. 2005). Recent years leled the introduction of the third generation of drugs,
also brought attempts to combined acceleration and namely paclitaxel (Johnson et al. 1996; Herscher et al.
hyperfractionation in less demanding regimen, such 1998), docetaxel (Millward et al. 1996; Mauer et al.
412 B. Jeremić et al.

1998), vinorelbine (LeChevalier et al. 1994; Masters days 1 and 29) and vinblastine (5 mg/m2, days 1, 8,
et al. 1998), gemcitabine (Manegold et al. 1997; 15, 22 and 29). Radiation therapy to a total dose of
Vokes et al. 1998), irinotecan (Fukuoka et al. 1992; 60 Gy in 30 fractions was given in both arms and
Oshita et al. 1997) and topotecan (Lynch et al. 1994; began on day 50 in the combined-modality arm. The
Perez-Soler et al. 1996). In most recent years, a addition of chemotherapy did not impair the ability to
number of new chamotherapy agents as well as tar- deliver radiation therapy; 88% of patients in the
geted agents have been introduced into the clinical combined-modality arm and 87% of patients on the
research in this disease, with the latter two being a radiation therapy alone arm completed radiation
subject of separate chapters of the book. therapy per protocol. Although there were no treat-
ment-related deaths on either arm, the addition of
chemotherapy increased the number of hospital
3 Neoadjuvant (Induction) admissions for vomiting (5% vs. 0%) and infection
Chemotherapy Followed (7% vs. 3%). In the initial report (Dillman et al.
by Radiation Therapy 1990), induction chemotherapy improved median
survival (13.8 vs. 9.7 months, p = 0.0066) and 3-year
Major aims of this type of combined radiation therapy survival of patients treated with radiochemotherapy
and chemotherapy are (1) to decrease tumor burden, (23% vs. 11%). Seven year follow-up of that study
which may permit delivery of radiation therapy to a confirmed superiority of induction chemotherapy
reduced tumor volume and (2) to combat micromet- (median survival, 13.7 vs. 9.6 months, p = 0.012)
astatic disease, believed to be present at the time of over radiation therapy alone (Dillman et al. 1996).
starting treatment. Increased drug delivery with less The Cancer and Leukemia Group B experience was
overall toxicity (especially those expected to occur subsequently been confirmed by other modern cis-
within the radiation therapy treatment field, i.e. platin-based trials (Table 1).
pneunomitis and esophagitis) is also one of the pos- The Radiation Therapy Oncology Group/Eastern
sible advantages if compared to concurrent adminis- Cooperative Oncology Group trial was a trial which
tration. Potential disadvantages of induction treatment randomized 458 patients with favourable prognosis
include (1) prolonged overall treatment time, (2) (good performance status, minimal weight loss)
excessive toxicity due to chemotherapy preventing or locally advanced nonsmall cell lung cancer to receive
delaying the delivery of full radiation therapy dose, either once daily radiation therapy to 60 Gy in 2 Gy
(3) chemotherapy-induced tumor cell resistance fractions or the same radiation therapy with two
resulting in reduced radiation therapy efficacy, as well cycles of induction cisplatin and vinblastine (Sause
as (4) accelerated tumor clonogen repopulation, et al. 1995), or the radiation therapy given twice daily
expected also to occur during the chemotherapy phase (1.2 Gy b.i.d.) to a total dose of 69.6 Gy. The median
of the combined treatment (Byhardt et al. 1998; survival was statistically superior (p = 0.03) for the
Byhardt 1999). Several phase III trials have demon- combined modality arm (13.8 months) versus either
strated a survival benefit for induction chemotherapy, the standard radiation therapy arm (11.4 months), or
confirmed with recent updates providing a long-term the twice daily radiation therapy arm (12.3 months).
data. Prolonged follow up of this study reconfirmed supe-
The Cancer and Leukemia Group B 8433 was the riority of combined-modality therapy. However, long-
landmark trial comparing sequential radiochemo- term survival rates remained less than 10% (Sause
therapy versus radiation therapy alone for the treat- et al. 2000).
ment of locally advanced nonsmall cell lung cancer French experience was provided in a phase III trial
(Dillman et al. 1990). Between 1984 and 1987, 155 with radiation therapy alone versus combined radia-
patients with clinical or surgical T3 or N2 and M0 tion therapy-chemotherapy (Le Chevalier et al. 1991).
nonsmall cell lung cancer were treated with induction In this trial, 353 patients with unresectable locally
chemotherapy followed by radiation therapy or radi- advanced squamous cell or large cell lung carcinoma
ation therapy alone. All patients had a good perfor- were randomized to either radiation therapy alone
mance status and minimal weight loss. Induction (65 Gy in 2.5 Gy fractions) or three monthly cycles of
chemotherapy consisted of cisplatin (100 mg/m2, cisplatin-based chemotherapy followed by the same
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 413

Table 1 Stage III nonsmall-cell lung cancer at University Hospital, Kragujevac, Serbia
Study Year Phase N Hfx RT (bid) CHT MST (mos) Survival
3 yr (%) 4 yr (%) 5 yr (%)
1 1995 III 61 64.8 – 8 7 7 5
56 64.8 CE 13 16 16 16
52 64.8 CE 18 23 21 21
2 1996 III 66 69.6 – 4 11 9
65 69.6 CE/d 22 23 23
3 1998 II 41 69.6 CE/d/w 25 34 29 29
4 2001 III 98 69.6 CE/d 20 29 29 20
97 69.6 CE/d/w 25 34 23 23
5 2005 II 64 67.6 CT/d 28 37 28 26
Hfx RT (bid) hyperfractionated radiation therapy, CHT chemotherapy, MST median survival time, C carboplatin, E etoposide,
T paclitaxel, d daily, w weekend

radiation therapy regimen and followed again by the latter is given alone. While this certainly holds
the same chemotherapy. A significant decrease in true for short-term survival, long-term survival figures
distant metastases for the combined-modality arm was remained to be dismal, indicating that only modest
observed and the median (12.0 vs. 10.0 months) and improvements in long-term survival have been
2-year survival rates (21% vs. 14%, p = 0.02) were achieved. Contrary to these, several smaller random-
improved as well (Le Chevalier et al. 1992). Long- ized trials have failed to confirm a survival benefit for
term follow up provided, unfortunately, more sobering the addition of induction chemotherapy, although low
facts: only 8% of patients had continued local control statistical power of these studies may have prevented
at 5 years (Arriagada et al. 1997), while 5-year sur- them to detect small differences in survival (Mattson
vival rates remained poor at 6 and 3% (for the two et al. 1988; Morton et al. 1991; Crino et al. 1993;
arms), likely a consequence of the high rate of local Planting et al. 1996). Three large meta-analyses have
failure on both arms. An intriguing question remained demonstrated a small but consistent survival benefit
unanswered: was improvement in the distant metas- for the addition of induction chemotherapy to radia-
tasis control a consequence of starting the treatment tion therapy for locally advanced nonsmall cell lung
with induction chemotherapy or it was a mere reflec- cancer (Non-small Cell Lung Cancer Collaborative
tion of higher total doses being given (post-radiation Group 1995; Marino et al. 1995; Pritchard and
therapy chemotherapy was also given)? Anthony 1996).
The Medical Research Council also performed a Since it became obvious that induction chemo-
trial which randomized 447 eligible patients with good therapy followed by conventionally fractionated,
performance status and localized, inoperable nonsmall radical radiation therapy unequivocally brings an
cell lung cancer to receive either radiation therapy increase in the locoregional failures, investigators
alone or cisplatin-based induction chemotherapy fol- made efforts to overcome this big clinical problem by
lowed by the same radiation therapy (Cullen et al. offering more intensive latter (radiation therapy) part
1997). On both arms, the median radiation therapy dose of the combined treatment. By doing so, Clamon et al.
in both arm was rather low, 50 Gy. The median survival (1999) compared then standard induction chemo-
time was improved with the addition of chemotherapy therapy consisting of cisplatin/vinblastine followed
(13.0 vs. 9.9 months, p = 0.056), although this dif- by standard radiation therapy (60 Gy in 30 daily
ference was of borderline (in)significance. fractions). Radiation therapy was given either with or
What these trials have demonstrated is that the without concurrently given weekly 100 mg/m2 of
addition of platinum-based induction chemotherapy carboplatin as radiosensitizer (enhancer). No differ-
to radiation therapy results in improved survival over ence in overall survival (the median survival time:
that obtained with the same radiation therapy when 13.4 vs. 13.5 months; 4-year survival: 13% vs. 10%;
414 B. Jeremić et al.

p = 0.74) was found between the radiosensitized and Cancer and Leukemia Group B (Clamon et al. 1999;
non-radiosensitized groups of patients. These results Vokes et al. 2002; Akerley et al. 2005; Socinski et al.
showed that induction cisplatin/vinblastine followed 2008) showed that there is no compensation for insuf-
by conventionally fractionated radical radiation ther- ficient start (i.e. with chemotherapy). Other attempts,
apy and concurrent chemotherapy that does not nec- such as the use of three daily fractions of radiation
essarily obtain good and consisting results, being therapy (Belani et al. 2005), also proved to be ineffec-
inferior in the study of Clamon et al. (1999) to what tive. In that trial which compared standard fractionation
was expected from previous two studies (Dillman radiation therapy versus hyperfractionated accelerated
et al. 1990; Sause et al. 1995). They have shown that radiation therapy, all patients in both arms received
even when sensitized by carboplatin, standard fraction induction chemotherapy with carboplatin/paclitaxel.
radiation therapy can not compensate for accelerated Concurrent chemotherapy was not used during the
proliferation of surviving tumor clonogens which radiation therapy course. Unfortunately, the study
occur during the induction (chemotherapy) phase of did not meet its accrual goals and was closed early;
treatment. Furthermore, the results of the study of nonetheless 111 patients were analyzed and the
Clamon et al. (1999) were not different from those results suggest a slight though statistically insignificant
obtained by hyperfractionated radiation therapy alone advantage to hyperfractionated accelerated radiation
in the Radiation Therapy Oncology Group/Eastern therapy (median survival and 2- and 3-year actuarial
Cooperative Oncology Group study 8808 (Sause et al. survival: 22.2 months, 48 and 20% vs. 13.7 months, 33
1995) or the same hyperfractionated radiation therapy and 15%, respectively) (Belani et al. 2005). All in all,
(69.6 Gy using 1.2 Gy twice-daily) in the study of whatever you do after you start with chemotherapy,
Jeremic et al. (1996). failure is inevitable and comes fast. With this approach,
More recently, in an attempt to intensify the second you can only achieve more toxicity (Vokes et al. 2002;
part (i.e. radiation therapy) of the combined treatment Socinski et al. 2008) and even if you use the modern
even more, Vokes et al. (2002) reported on randomized radiation therapy tools such as three-dimensional
phase II study of Cancer and Leukemia Group B (9431) radiation therapy and attempt treatment intensification
which used two cycles of induction chemotherapy by escalating the total dose, again, one cannot achieve
(cisplatin/gemcitabine or cisplatin/paclitaxel or cis- better outcome. Indeed, impressive 12% mortality in
platin/vinorelbine) followed by the two cycles of the the most recent CALGB attempt (Socinski et al. 2008)
same chemotherapy concurrently with conventionally to combine induction chemotherapy with subsequent
fractionated radical radiation therapy (66 Gy) in 175 radiation therapy and concurrent chemotherapy led
patients with unresectable stage III nonsmall cell lung investigators to early stopping the trial.
cancer. Response rates were 74, 67, and 73% for the Underlying radiobiological principles of this
three arms, respectively. While the median survival observation can easily be materialized in a recently
time for all patients was 17 months, 3-year survival identified fact of never-achieved improvement in
rates for the three groups were 28, 19, and 23%, local control in locally advanced nonsmall cell lung
respectively. Although authors concluded that the use cancer treated with induction chemotherapy followed
of concurrent radiochemotherapy could have led to the by either conventionally fractionated radical radiation
improvement in outcome when compared to previous therapy or even stronger, radiochemotherapy. El
Cancer and Leukemia Group B experience with Sharouni et al. (2003) has compared CT scans pre-
induction treatments (Dillman et al. 1990, 1996; and post-induction chemotherapy, with emphasis on
Clamon et al. 1999), it did not improve treatment out- the time from the last induction chemotherapy cycle
come when one considers other combined modality to the time of radiation therapy treatment planning CT
options such as concurrent radiation therapy and che- was actually done. That way they have been able to
motherapy. Several studies of the similar design only measure an increase in gross tumour volumes (GTVs)
reconfirmed these observations. They have clearly and subsequently define volume doubling times.
shown that ANY intensification of the latter/major part During the waiting period (for the planning CT scan
of the combined treatment approach via concurrent and start of radiation therapy), a total of 41% of all
radiochemotherapy is not effective, once you have tumours became incurable, with the ratio of GTVs
started with induction chemotherapy. Several studies of being in the range of 1.1–81.8! Tumour doubling
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 415

times ranged 8.3–171 days, with the median of III studies investigated this issue (Soresi et al. 1988;
29 days. When translated into more clinical language, Schaake-Koning et al. 1992; Trovo et al. 1992;
these findings clearly say that even if one may have Blanke et al. 1995; Jeremic et al. 1995, 1996; Bonner
thought (due to insufficient CT-based imaging) that et al. 1998; Groen et al. 2004; Ball et al. 1999).
response occurs (and that it matters), there is actually Relatively low total radiation therapy dose (Soresi
an opposite development, with surviving tumour et al. 1988; Trovo et al. 1992) and chemotherapy
clonogens repopulating fast, leading tumours to being given in an insufficient total dose (Soresi et al.
regrow to the state of incurability. Recent confirma- 1988) may have been among reasons for such
tion of these observations came from Bozcuk et al. observation. All three positive studies used protracted
(2010) who attempted to correlate the benefit from chemotherapy dosing. While an European Organiza-
neoadjuvant chemotherapy before radiotherapy in tion for Research and Treatment of Cancer study
non-small cell lung cancer using a meta-analytical (Schaake-Koning et al. 1992) tested both daily and
approach with meta-regression analysis. They have weekly cisplatin with split-course radiation therapy,
used thirteen randomized clinical trials to date, showing superior outcome for daily cisplatin/radia-
encompassing 2776 patients. Time to radiotherapy tion therapy, Jeremic et al. first used biweekly and
was inversely associated with the benefit from neo- weekly (Jeremic et al. 1995) and then daily (Jeremic
adjuvant chemotherapy at 2 (p = 0.050) and 3 years et al. 1996) carboplatin/etoposide with hyperfrac-
(p = 0.093). This meta-analysis highlighted the tionated radiation therapy doses of 64.8 (Jeremic et al.
importance of shorter time to radiotherapy to maxi- 1995) and then 69.6 Gy (Jeremic et al. 1996). The
mize nonsmall cell lung cancer patients’ survival. best results were obtained with low-dose daily che-
motherapy given during the hyperfractionated radia-
tion therapy course, with 4–5 year survival rates
4 Concurrent Radiochemotherapy being at the order of 20% (Jeremic et al. 1995, 1996).
Survival advantage in these three studies was a con-
This combined modality approach denotes the admin- sequence of an advantage at local tumour level,
istration of both modalities at the same time, meaning confirming radiobiological expectations that low-dose
that chemotherapy is given during the course of radical, daily chemotherapy may have acted synergistically
curative radiation therapy. A number of variations with radiation therapy, and enhanced its effects on
exist, including chemotherapy being administered on a local tumour level. Also as expected, no influence on
3-weekly basis, bi-weekly, weekly, or daily, although distant metastasis control was noted. Ulutin et al.
concurrent radiation therapy–chemotherapy employ- (2000) designed a three-armed study to compare the
ing third generation drugs (e.g. paclitaxel) also wit- effects of sequential and concurrent radiation ther-
nessed administration of the drug twice or trice weekly. apy–chemotherapy in locally advanced non-small cell
Whatever was the design of concurrent radiation ther- lung cancer. Each treatment arm consisted of 15
apy–chemotherapy, its main aim is to address the issue patients with histologically confirmed stage III non-
of locoregional and distant disease at the same time, small cell lung cancer. In group 1, the main treatment
from the outset of the treatment as intensively as pos- approach was split-course radiation therapy alone. In
sible. This, unfortunately, may lead to increased tox- group 2, 6 mg/m2 of cisplatin was applied daily and
icity (mostly acute) which may require dose reductions concurrently with split-course radiation therapy. In
or treatment interruptions, both adversely influencing group 3, two cycles of etoposide, ifosfamide, and
treatment outcome. On the other side, with this cisplatin chemotherapy, which ended 3 weeks before
approach three of radiobiological premises, namely split-course radiation therapy, was applied. Overall
spatial cooperation, independent cell kill and syner- response rates were 40, 66, and 53% in groups
gistic action, as postulated by Steel and Peckham 1–3, respectively. Median survival was 10, 11, and
(1979), can be exploited. 10 months for groups 1–3, respectively. Recently,
Concurrent radiation therapy–chemotherapy was another study from Turkey (Cakir and Egehan 2004)
compared to radiation therapy alone, the former provided additional evidence that concurrent radiation
aiming mostly on an improvement at local tumour therapy (64 Gy in 32 daily fractions) and cisplatin
control. A number of prospective randomized phase (20 mg/sqm, days 1–5, weeks 2 and 6) offers survival
416 B. Jeremić et al.

advantage over the same radiation therapy alone cancer, stratified by performance status, stage, and
(3-year survival, 10% vs. 2%). Combined treatment institution, were randomly assigned to two arms
approach also offered better locoregional control consisting of docetaxel 40 mg/sqm and cisplatin
(p = 0.0001) and disease-free survival (p = 0.0006). 40 mg/sqm on days 1, 8, 29, and 36) or mitomycin,
Most recently, West Japan Thoracic Oncology Group vindesine, and cisplatin chemotherapy with concur-
(Yamamoto et al. 2010) presented the data from a rent thoracic radiation therapy. Two hundred patients
phase III trial of concurrent thoracic radiation therapy were allocated into either the docetaxel and cisplatin
(WJTOG0105) which was conducted to compare or mitomycin, vindesine, and cisplatin arm. The
third-generation chemotherapy with second-genera- survival time at 2 years, a primary end point, was
tion chemotherapy in patients with unresectable favourable to the docetaxel and cisplatin arm
stage III nonsmall-cell lung cancer. Eligible patients (p = 0.059 by a stratified log-rank test as a planned
received the following treatments: A (control), four analysis and p = 0.044 by an early-period, weighted
cycles of mitomycin (8 mg/sqm on day 1)/vindesine log-rank as an unplanned analysis). There was a trend
(3 mg/sqm on days 1, 8)/cisplatin (80 mg/msqm on toward improved response rate, 2-year survival rate,
day 1) plus thoracic radiation therapy 60 Gy median progression-free time, and median survival in
(treatment break for 1 week); B, weekly irinotecan the docetaxel and cisplatin arm (78.8, 60.3%, 13.4,
(20 mg/sqm)/carboplatin (area under the plasma and 26.8 months, respectively) compared with the
concentration–time curve, 2) for 6 weeks plus tho- mitomycin, vindesine, and cisplatin arm (70.3, 48.1%,
racic radiation therapy 60 Gy, followed by two 10.5, and 23.7 months, respectively), which was not
courses of irinotecan (50 mg/sqm on days 1, 8)/car- statistically significant (p [ 0.05). Grade 3 febrile
boplatin (area under the plasma concentration–time neutropenia occurred more often in the mitomycin,
curve 5 on day 1); C, weekly paclitaxel (40 mg/sqm)/ vindesine, and cisplatin arm than in the docetaxel and
carboplatin (area under the plasma concentration– cisplatin arm (39% vs. 22%, respectively; p = 0.012),
time curve, 2) for 6 weeks plus thoracic radiation and grade 3–4 radiation esophagitis was likely to be
therapy 60 Gy, followed by two courses of paclitaxel more common in the docetaxel and cisplatin arm than
(200 mg/sqm on day 1)/carboplatin (area under the in the mitomycin, vindesine, and cisplatin arm (14%
plasma concentration–time curve 5 on day 1). The vs. 6%, p = 0.056). Authors concluded that the
median survival time and 5-year survival rates were docetaxel and cisplatin chemotherapy combined with
20.5, 19.8, and 22.0 months and 17.5, 17.8, and concurrent thoracic radiation therapy is an alternative
19.8% in arms A–C, respectively. While no signifi- to mitomycin, vindesine, and cisplatin chemotherapy
cant differences in overall survival were apparent for patients with locally advanced nonsmall cell lung
among the treatment arms, noninferiority of the cancer.
experimental arms was not achieved. The incidences Additional and important observation coming from
of grade 3–4 neutropenia, febrile neutropenia, and clinical trials data accumulated over the years is that
gastrointestinal disorder were significantly higher in those studies/arms which used high-dose chemother-
arm A than in arm B or C (p \ 0.001). Chemotherapy apy (mimicking classic chemotherapy administration
interruptions were more common in arm B than in and necessarily given with more split between the
arm A or C. Arm C was equally efficacious and consecutive chemotherapy cycles) concurrently with
exhibited a more favourable toxicity profile among radiation therapy neither observed any impact on
three arms. This study confirmed effectiveness of distant metastasis control, nor did so on local level.
third-generation chemotherapy and concurrent radia- Beside the overall treatment success, another advan-
tion therapy in this setting. In a similar study, also tage of low-dose concurrent chemotherapy over high-
coming from Japan, Segawa et al. (2010) reported on dose chemotherapy and concurrent radiation therapy
a phase III trial comparing docetaxel and cisplatin is that the former type of concurrent radiochemo-
combination chemotherapy with mitomycin, vinde- therapy leads to less high-grade acute toxicity and,
sine, and cisplatin combination chemotherapy with consequently, better treatment compliance, less
concurrent thoracic radiotherapy in locally advanced treatment interruptions, which influence on treatment
non-small-cell lung cancer. Patients age 75 years or outcome (Cox et al. 1993). Recently, Harada et al.
younger with locally advanced nonsmall cell lung (2009) retrospectively compare the survival and
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 417

toxicities associated with radiochemotherapy using enhancement of tumour response may have occurred.
full-dose and weekly regimens in patients with stage In the low-dose (daily) chemotherapy arms of the
III non-small cell lung cancer. Fifty-nine patients concurrent studies, however, it seems unlikely that
were enrolled; 36% of the patients were treated with independent cell kill occurred (and if so, then to a
full-dose regimens and 64% with weekly regimens. In much lesser degree due to lower daily and total doses
both univariate and multivariate analyses, treatment of chemotherapy), leaving, thus, enhancement of
with weekly regimens was associated with a better tumour response as the only and likely alternative.
overall survival than that with full-dose regimens In order to compare induction chemotherapy fol-
(2-year survival rates: 75% for weekly regimens vs. lowed by radical radiation therapy to concurrent
41% for full-dose regimens). The toxicities and radiation therapy and chemotherapy, several clinical
compliance in the two groups were comparable. trials were performed. The very first full publication
Weekly regimens exhibited more favourable overall came from The Cancer and Leukemia Group B
survival as compared to full-dose regimens in this (Clamon et al. 1999) who reported on a randomized
retrospective study. These results support previous phase II trial where induction chemotherapy (cis-
observations that low-dose daily chemotherapy platin–vinblastin) was followed with either radical
(Schaake-Koning et al. 1992; Jeremic et al. 1995, radiation therapy alone and then followed with four
1996, 2005) provide good treatment results including additional chemotherapy cycles (regimen 1) or with
the toxicity which is lower than that observed when concurrent radical radiation therapy and six weekly
high-dose radiation therapy and concurrent high-dose doses of carboplatin (regimen 2). The additional four
chemotherapy are administered in this setting. As cycles of vinblastine and cisplatin were completed by
such an example, results of studies coming from the 34% of patients; the radiation therapy and concurrent
University Hospital, Kragujevac, Seriba are chrono- carboplatin was completed by 70% of patients. Grade
logically outlined in Table 1. 3 or 4 granulocytopenia occurred in 53% of patients
on regime 1 versus 17% on regime 2 (p \ 0.003);
grade 3 or 4 nausea/vomiting occurred in 20% of
5 Neoadjuvant (Induction) those on regime 1 versus 7% on regimen 2
Chemotherapy Followed (p = 0.175). Response rates and survival were similar
by Radiation Therapy Versus for the two regimens, with approximately 30% of
Concurrent Radiochemotherapy patients surviving at 2 years. Nest study originated in
Japan were Furuse et al. (1999, 2000) reported on a
The studies using induction chemotherapy followed study of the West Japan Lung Cancer Group which
by radical radiation therapy showed a survival compared mitomycin, cisplatin, and vindesine che-
advantage for the combined approach owing to the motherapy given as either induction followed by
improvement in the distant metastasis control. It is a continuous course radical radiation therapy (56 Gy)
finding opposite to that of the studies using concurrent with the same mitomycin, cisplatin, and vindesine
radiation therapy–chemotherapy, which unequivo- given concurrently with split-course radiation therapy
cally showed improvement in survival owing to of the same total dose. First publication showed
the improvement in locoregional tumour control. superior results (the median survival time, 16.5 vs.
Considered from the standpoint of exploitable 13.3 months; 5-year survival, 16% vs. 9%;
mechanisms of combined radiation therapy and che- p = 0.039) for concurrent radiation therapy–chemo-
motherapy (Steel and Peckham 1979), the induction therapy (Furuse et al. 1999). Subsequent data analysis
regimens enabled the therapeutic benefit due to spatial showed that an improvement in local tumour control
cooperation only. Neither independent cell kill nor (median time, 10.6 vs. 8.0 months; 5-year, 34% vs.
enhancement of tumour response could be noted 20%; p = 0.0462) is the reason for an improvement
because there was no significant difference in loco- in survival (Furuse et al. 2000). More recently, Curran
regional tumour control, as one may expect if the two et al. (2000) and Komaki et al. (2000) reported on
mechanisms of action would have happened. On the Radiation Therapy Oncology Group 9410 study
other side, in concurrent studies, spatial cooperation which compared (1) induction chemotherapy fol-
did not work, while both independent cell kill and lowed by radiation therapy, same as Cancer and
418 B. Jeremić et al.

Leukemia Group B 8433 (Dillman et al. 1990) and days 1 to 78, followed by thoracic radiation therapy at
Radiation Therapy Oncology Group 8808/Eastern a dose of 66 Gy in 33 fractions (2 Gy per fraction and
Cooperative Oncology Group 4508 (Sause et al. five fractions per week) or the same radiation therapy
1995) with concurrent either (2) standard fraction (started on day 1) with two concurrent cycles of cis-
radiation therapy (60 Gy) and cisplatin/etoposide or platin 20 mg/sqm/d and etoposide 50 mg/sqm/d (days
(3) hyperfractionated radiation therapy (69.6 Gy) and 1–5 and days 29–33); patients then received consoli-
cisplatin/etoposide. Both standard radiation therapy– dation therapy with cisplatin 80 mg/sqm on days 78
chemotherapy and hyperfractionated radiation ther- and 106 and vinorelbine 30 mg/sqm/wk from days 78
apy–chemotherapy arm had better median survival to 127. There were six toxic deaths in the induction
time than the induction arm (17.0 vs. 16.0 vs. arm and 10 in the concurrent arm. Median survival
14.6 months), although only standard radiation ther- was 14.5 months in the induction arm and
apy–chemotherapy was statistically significantly bet- 16.3 months in the concurrent arm (p = 0.24). Two-,
ter than induction chemotherapy (Curran et al. 2000). 3-, and 4-year survival rates were better in the con-
Pattern of failure analysis showed that the best local current arm (39, 25, and 21%, respectively) than in
control was in the hyperfractionated radiation ther- the induction arm (26, 19, and 14%, respectively).
apy–chemotherapy arm, confirming indirectly the Esophageal toxicity was significantly more frequent
observations of Jeremic et al. (1995, 1996) that high- in the concurrent arm than in the induction arm (32%
dose hyperfractionated radiation therapy–chemother- vs. 3%). Although not statistically significant, differ-
apy is an advantageous approach. Furthermore and ences in the median, 2-, 3-, and 4-year survival rates
contrary to studies using low-dose chemotherapy which were observed were clinically meaningful, with
concurrently with high-dose radiation therapy, it was a trend in favor of concurrent radiation therapy–che-
shown again that high-dose chemotherapy bears a risk motherapy, suggesting that is the optimal strategy for
of exceptional acute toxicity when given with high- patients with locally advanced nonsmall cell lung
dose standard or hyperfractionated radiation therapy. cancer. Finally, Belderbos et al. (2007) reported on
This finding is not just limited to Radiation Therapy EORTC study in 158 patients which were randomised
Oncology Group 9410 but was also seen in other to receive two courses of (induction) Gemcitabine
similar studies (Byhardt et al. 1995; Lee et al. 1996; (1250 mg/sqm days 1, 8) and Cisplatin (75 mg/sqm,
Komaki et al. 1997) as well. The next study was day 2) prior to, or daily low-dose Cisplatin (6 mg/
aforementioned Turkish study of Ulutin et al. (2000), sqm) concurrent with radiotherapy, consisting of 24
designed as three-armed study (including the group 1 fractions of 2.75 Gy in 32 days, with a total dose of
was split-course radiation therapy alone) to compare 66 Gy. Acute haematological toxicity grade 3/4 was
the effects of sequential and concurrent radiation more pronounced in the induction arm (30% versus
therapy–chemotherapy in locally advanced non-small 6%), oesophagitis grade 3/4 more frequent in the
cell lung cancer. Each treatment arm consisted of 15 concurrent arm (5% versus 14%). Late oesophagitis
patients with histologically confirmed stage III non- grade 3 was 4% (induction and concurrent), pneu-
small cell lung cancer. In group 2, 6 mg/sqm of cis- monitis grade 3/4 14% (induction) and 18%
platin was applied daily and concurrently with split- (concurrent). Because of the poor power of the study
course radiation therapy. In group 3, two cycles of no significant differences in median survival time
etoposide, ifosfamide, and cisplatin chemotherapy, (induction, 16.2, concurrent, 165 months, respec-
which ended 3 weeks before split-course radiation tively), and 3-year overall survival (induction, 22%,
therapy, was applied. Overall response rates were 40, concurrent, 34%) could be detected.
66, and 53% in groups 1–3, respectively. Median Although discussed clinical trials have clearly
survival was 10, 11, and 10 months for groups 1–3, (though not always statistically significantly!) shown
respectively. In France, Groupe Lyon-Saint-Etienne that concurrent radiation therapy–chemotherapy may
d’Oncologie Thoracique-Groupe Français de be considered as standard treatment option in, at least
Pneumo-Cancérologie (Fournel et al. 2005) randomly favorable (good performance status, less pronounced
assigned 205 patients to receive either induction weight loss) patients with locally advanced nonsmall
chemotherapy with cisplatin (120 mg/sqm) on days 1, cell lung cancer, last decade was the time of contin-
29, and 57, and vinorelbine (30 mg/sqm/wk) from uous discussions in this field. While proponents of
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 419

concurrent radiation therapy–chemotherapy cited individual study. Pooled odds ratios for the objective
high level evidence, opponents were usually using response rate, relapse control rate, and toxic events
various explanations and excuses for not using it in were calculated using the Mantel–Haenszel estimate.
daily practice. Some of these were showing ineffi- Results confirmed that radiation therapy and concur-
ciency of administration matters at radiation oncology rent chemotherapy determined a statistically signifi-
departments, such as existing waiting lists and/or long cant increase in median survival time (16.3 vs.
time to organize CT-scanning for treatment planning, 13.9 months; pooled median ratios = 1.17, 95% CI
even in the leading institutions worldwide. To solve 1.09–1.26), response rate (64.0% vs. 56.3%; odds
major scientific and clinical question, but also to use ratio = 1.38, 95% CI 1.10–1.72), and tumor-relapse
the opportunity to solve some, if not all of then- control (odds ratio = 0.82,95% CI 0.69–0.97), though
existing uncertainties, three meta-analyses/systematic at the expense of increased hematological toxicity
reviews were performed. (neutropenia and thrombocytopenia) and non-hema-
O’Rourke et al. (2010) identified nineteen random- tological toxicity (nausea/vomiting, stomatitis, and
ised studies (totaling 2728 participants) of radiation esophagitis). Similar results were obtained from the
therapy and concurrent chemotherapy versus radiation sensitivity analysis of all Phase-III trials designed to
therapy alone. Radiation therapy and concurrent che- evaluate the primary end point of overall survival.
motherapy significantly reduced overall risk of death Subgroup analysis revealed that concurrent strategy
(HR 0.71, 95% CI 0.64–0.80; I(2) 0%; 1607 partici- was mainly associated with improved loco-regional
pants) and overall progression-free survival at any site control (odds ratio = 0.68, 95% CI 0.52–0.87). How-
(HR 0.69, 95% CI 0.58–0.81; I(2) 45%; 1145 partici- ever, no difference in progression-free survival is
pants). Incidence of acute oesophagitis, neutropenia shown. While careful interpretation of their conclu-
and anaemia were significantly increased with radia- sions is required because of potential bias, authors
tion therapy and concurrent chemotherapy. Six trials concluded that further improvements will be obtained
(1024 patients) of radiation therapy and concurrent by optimizing the conditions for a concurrent regimen.
chemotherapy versus induction chemotherapy and Finally, Aupérin et al. (2010) used updated indi-
radiation therapy included. A significant benefit of vidual patient data to address the same question.
chemotherapy was shown in overall survival (HR 0.74, Results from trials were combined using the stratified
95% CI 0.62–0.89; I(2) 0%; 702 participants). This log-rank test to calculate pooled hazard ratios. The
represented a 10% absolute survival benefit at 2 years. primary outcome was overall survival; secondary out-
More treatment-related deaths (4% vs. 2%) were comes were progression-free survival, cumulative
reported in the radiation therapy and concurrent che- incidences of locoregional and distant progression, and
motherapy arm without statistical significance (RR acute toxicity. Of seven eligible trials, data from six
2.02, 95% CI 0.90–4.52; I(2) 0%; 950 participants). trials were received (1,205 patients, 92% of all ran-
There was increased severe oesophagitis with radiation domly assigned patients). Median follow-up was
therapy and concurrent chemotherapy (RR 4.96, 95% 6 years. There was a significant benefit of radiation
CI 2.17–11.37; I(2) 66%; 947 participants). therapy and concurrent chemotherapy on overall sur-
Liang et al. (2010) investigated whether current vival (pooled hazard ratio, 0.84; 95% CI 0.74–0.95;
clinical trials can clarify this schedule and offer further p = 0.004), with an absolute benefit of 5.7% (from 18.1
bases for clinical decision making. They performed a to 23.8%) at 3 years and 4.5% at 5 years. For pro-
systematic review of 11 trials (2,043 patients; concur- gression-free survival, the pooled hazard ratios was
rent—1,019, induction—1,024) that compared radia- 0.90 (95% CI 0.79–1.01; p = 0.07). Radiation therapy
tion therapy and concurrent chemotherapy with and concurrent chemotherapy decreased locoregional
induction chemotherapy followed with radiation ther- progression (pooled hazard ratios, 0.77; 95% CI 0.62–
apy in advanced nonsmall cell lung cancer patients. 0.95; p = 0.01); its effect was not different from that of
Primary end point was overall survival. Pooled median induction treatment on distant progression (pooled
ratios and progression-free-survival ratios for median hazard ratios, 1.04; 95% CI 0.86–1.25; p = 0.69).
survival and progression-free survival were calculated Radiation therapy and concurrent chemotherapy
using the weighted sum of the log ratio of pooled increased acute esophageal toxicity (grade 3–4) from 4
median ratio and progression-free-survival ratios of to 18% with a relative risk of 4.9 (95% CI 3.1–7.8;
420 B. Jeremić et al.

p \ 0.001). There was no significant difference While Jeremic et al. (1998) tested the addition of
regarding acute pulmonary toxicity. Overall conclu- weekend carboplatin/etoposide to concurrent hyper-
sion of this meta-analysis was that radiation therapy fractionated radiation therapy (69.6 Gy) and low-dose
and concurrent chemotherapy, as compared with daily carboplatin/etoposide in phase II study leading
induction chemotherapy and radiation therapy, to promising median survival time of 29 months and
improved survival of patients with locally advanced 5-year survival in 25%, the results of their subsequent
nonsmall cell lung cancer, primarily because of a better prospective randomized trial showed no advantage for
locoregional control, but at the cost of manageable weekend chemotherapy when compared to no week-
increased acute esophageal toxicity. end chemotherapy (the median survival time, 22 vs.
With three meta-analyses the story of superiority 20 months; 5-year survival, 23% vs. 20%; p = 0.57)
of radiation therapy and concurrent chemotherapy (Jeremic et al. 2001). In their continuous efforts to
over the induction chemotherapy followed by radical address important questions in this disease and the
radiation therapy seems to finally be over. Further setting of radiation therapy and concurrent chemo-
studies should attempt to optimize concurrent therapy, Jeremic et al. (2005) (Table 1) recently
approach only as it seems that there are still, however, pioneered a combined modality approach consisting
a number of unsolved issues. of hyperfractionated radiation therapy and concurrent
While we are embarking on these efforts, one par- low-dose daily paclitaxel and carboplatin. In order to
ticular issue could easily be solved. Radiation therapy increase likelihood of successfully combating accel-
and concurrent cemotherapy is still labelled as CHE- erated proliferation of tumour clonogens, they have
MORADIATION. More appropriate, however, termi- adapted their initial standard regimen (69.6 Gy in 58
nology for a combination of radiation therapy and fractions of 1.2 Gy given b.i.d) in 6 weeks to 67.6 Gy
concurrent chemotherapy is RADIOCHEMOTHER- in 52 fractions of 1.3 Gy given also bid, but in a
APY. If one gives 65–70 Gy of radiation therapy 5-week total treatment time, saving approximately 1
and either daily low-dose chemotherapy or weekly week. Paclitaxel was given in a daily dose of 10 mg/
chemotherapy or even more protracted chemotherapy sqm, while carboplatin was given in a daily dose of
(i.e. cisplatin, 100 mg/sqm, day 1 and etoposide, 25 mg/sqm. In 64 patients with stage III nonsmall cell
120 mg/sqm, days 1–3, q 3 weeks) how this can be lung cancer very promising median survival time of
called chemoradiation? Perhaps chemoradiation was 28 months and a 5-year survival rate of 26% were
more appropriate term in ancient times, when one obtained, accompanied with low incidence of high-
started with chemotherapy. Since this sequencing is grade toxicity. These results again reconfirmed
inferior to one including radiation therapy from the effectiveness and low toxicity of hyperfractionated
onset, the term ‘‘chemoradiation’’ should be put into the radiation therapy and concurrent low-dose daily
museum of history of failures. Radiation therapy is chemotherapy.
THE mainstay of the treatment and chemotherapy is Last decade witnessed a particular approach aimed
used to support these effects, this or that way. There- to optimize radiation therapy and concurrent chemo-
fore, RADIOCHEMOTHERAPY should be preferen- therapy (Table 2). By adding more chemotherapy
tially used as the term appropriate for all concurrent after the end of concurrent radiation therapy and
regimens, deemed today as standard of treatment in chemotherapy, i.e. posterior, adjuvant, consolidation
locally advanced nonsmall cell lung cancer, as recently chemotherapy, better treatment of microscopic dis-
shown by tree meta analysis (O’Rourke et al. 2010; ease is sought. Results of several available phase II
Liang et al. 2010; Aupérin et al. 2010). studies are given in table. First of such reports seem to
be that of Lau et al. (2001) who tried to address the
issue of somewhat poorer distant metastasis control
6 Optimization of Concurrent by increasing the dose of chemotherapy. They have
Radiochemotherapy used concurrent radiation therapy (61 Gy) and che-
motherapy consisting of twice weekly paclitaxel for
In order to optimize combined modality approach in 6 weeks and once weekly carboplatin for 6 weeks.
patients with locally advanced nonsmall cell lung Two cycles of consolidation paclitaxel and carbo-
cancer several new attempts have been undertaken. platin were offered to patients who achieved a
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 421

Table 2 Locally advanced nonsmall cell lung cancer consolidation chemotherapy studies
Study Year Concurrent RT-CHT Consolidation CHT MST (mo) Survival
Lau et al. 2001 RT/TC TC 9 2 17 40% (2 yr)
Albain et al. 2002 RT/PE PE 9 2 15 17% (5 yr)
Gandara et al. 2003 RT/PE DOC 9 3 26 29% (5 yr)
Sakai et al. 2004 RT/DC DC 9 2 27 61% (2 yr)
Sekine et al. 2006 RT/PV DOC 9 3 30 43% (3 yr)
Hanna et al. 2008 RT/PE No 23.2 26% (3 yr)
RT/PE DOC 9 3 21.2 27% (3 yr)
RT radiation therapy, CHT chemotherapy, MST median survival time, T paclitaxel, C carboplatin, P cisplatin, E etoposide, DOC
docetaxel, V vinorelbine

complete response (CR), partial response (PR), or cisplatin and vinorelbine in patients with unresectable
stable disease (SD). The median survival time was stage III non-small cell lung cancer. The median
17 months and 2-year actuarial survival rate was progression-free survival was 12.8 (CI 10.2–15.4)
40%. Another report comes also from Southwest months, and median survival was 30.4 (CI 24.5–36.3)
Oncology Group phase II study by Albain et al. months. The 1-, 2-, and 3-year survival rates were
(2002) which used two cycles of cisplatin/etoposide 80.7, 60.2, and 42.6%, respectively. The most com-
concurrently with conventionally fractionated 45 Gy mon reason for discontinuation was pneumonitis,
in pathologic Stage IIIB nonsmall cell lung cancer. which developed in 14 (24%) of the 59 patients.
In the absence of progressive disease, additional During consolidation therapy, grade 3 or 4 neutro-
16 Gy was administered with two additional cycles of penia, esophagitis, and pneumonitis developed in
cisplatin/etoposide. The median survival time was 51, 2, and 4 patients, respectively. A total of four
15 months and 3- and 5-year survival was 17 and patients died of pneumonitis.
15%, respectively. However, grade 4 neutropenia was Promising results of several phase II studies dis-
observed in 32% patients, grade 3–4 anemia in 28% cussed above clearly called for verification in a phase
patients, and grade 3–4 esophagitis in 20% patients. II study design. In such an attempt, Hosier Oncology
More recently, and quite encouragingly, the South Group (Hanna et al. 2008) enrolled eligible patients
West Oncology Group reported a trial in which con- with stage IIIA or IIIB nonsmall cell lung cancer,
current cisplatin/etoposide/radiation therapy was fol- baseline performance status of 0–1, forced expiratory
lowed by three cycles of adjuvant high-dose docetaxel volume in 1 s C 1 L, and less than 5% weight loss.
(Gandara et al. 2003). The median survival in this Patients received cisplatin 50 mg/sqm intravenously
phase II study was an extremely impressive (IV) on days 1, 8, 29, and 36 and etoposide 50 mg/
26 months, and this has become the basis for ongoing sqm IV on days 1–5 and 29–33 concurrently with
South West Oncology phase III studies. Sakai et al. chest radiation therapy to 59.40 Gy. Patients who did
(2004) also reported on a phase II study which not experience progression were randomly assigned to
employed bi-weekly docetaxel and carboplatin with docetaxel 75 mg/sqm IV every 21 days for three
concurrent radiation therapy (60 Gy in 30 daily cycles versus observation. Grade 3–5 toxicities during
fractions) followed by consolidation chemotherapy docetaxel included febrile neutropenia (10.9%) and
with docetaxel plus carboplatin in patients with stage pneumonitis (9.6%); 28.8% of patients were hospi-
III unresectable nonsmall cell lung cancer. Among 32 talized during docetaxel (vs. 8.1% in observation
evaluable patients, impressive response rate of 91% arm), and 5.5% died as a result of docetaxel.
was obtained. The median survival time was The median survival time for all patients (n = 203)
27 months and a 2-year survival was 61%. High- was 21.7 months; the median survival time was
grade toxicity was low. Most recently, another phase 21.2 months for docetaxel arm compared with
II study from Japan (Sekine et al. 2006) provided the 23.2 months for observation arm (p = 0.883). Based
data on the use of docetaxel consolidation therapy on these results, authors concluded that consolidation
following thoracic radiation therapy and concurrent docetaxel after radiation therapy and concurrent
422 B. Jeremić et al.

cisplatin/etoposide results in increased toxicities but with the patterns of failure. While these studies pre-
does not further improve survival compared with sented very detailed pattern of failure in general, this
radiation therapy and concurrent cisplatin/etoposide was done for the whole time period of the study
alone in patients with stage III inoperable nonsmall (treatment plus follow-up). This way we only learned
cell lung cancer. Finally, Kelly et al. (2008) reported about the total patterns of failure and not about which
on a study that used targeted agents in consolidation type of failure was observed when, i.e. after concur-
phase. Untreated patients with stage III nonsmall cell rent or after consolidation part, and particularly in
lung cancer, a performance score of 0–1, and ade- which patients after the concurrent part, although
quate organ function were eligible. All patients some studies mandated consolidation chemotherapy
received cisplatin 50 mg/sqm on days 1 and 8 plus in non-progressing patients.
etoposide 50 mg/sqm on days 1–5, every 28 days for One may, therefore, ask why exact pattern of
two cycles with concurrent thoracic radiation (1.8- to failure is important? It is important from several
2-Gy fractions per day; total dose, 61 Gy) followed standpoints, some of which are briefly outlined here.
by three cycles of docetaxel 75 mg/sqm. Patients First, there are several types of patients after the ini-
whose disease did not progress were randomly tial (concurrent) part of radiochemotherapy and they
assigned to gefitinib 250 mg/day or placebo until can easily be separated regarding the response. While
disease progression, intolerable toxicity, or the end of it is extremely unlikely that those achieving a SD
5 years. An unplanned interim analysis conducted would benefit from the consolidation chemotherapy,
rejected the alternative hypothesis of improved sur- those with either a CR or a PR seems as likely can-
vival. The study, therefore, closed, and preliminary didates (although not all of them) to benefit from the
results were reported. The median survival time was consolidation chemotherapy. Separation, therefore, of
23 months for gefitinib (n = 118) and 35 months for pattern of failure occurring in likely (CR and PR) and
placebo (n = 125; two-sided p = 0.013). The toxic unlikely (SD) candidates could be used for further
death rate was 2% with gefitinib compared with 0% studies using similar design with respect to, e.g. eli-
for placebo. Authors concluded that in this unselected gibility criteria. Second, and more importantly,
population, gefitinib did not improve survival. among likely candidates (CR and PR) to benefit from
Decreased survival was a result of tumor progression consolidation chemotherapy, a distinction should be
and not gefitinib toxicity. This study showed not only made between those achieving CR and those
inefficiency of targeted agent gefitinib, but recon- achieving PR after concurrent radiochemotherapy.
firmed inefficiency of consolidation chemotherapy This is so, since different mechanisms (precisely,
approach in locally advanced nonsmall cell lung different location) of action of consolidation chemo-
cancer. therapy would be expected. In the CR patients, con-
These disappointing results largely stopped the use solidation chemotherapy would target microscopic
of consolidation chemotherapy in this disease. While disease both intrathoracically and extrathoracically,
reasons for such inefficiency may be multiple, it is while in the PR patients, it would have to deal with
challenging to disclose some basic aspects that may clinically overt intrathoracic disease and a micro-
have adversely influenced the outcome from the scopic one extrathoracically. It is obvious that pattern
onset. Indeed, by virtue of the intervention, this of failure in these two distinct groups of patients
approach has two parts, a concurrent one and a con- would then clearly show how and where consolidation
solidation one. In various studies, the same or dif- chemotherapy is actually acting and to what extent
ferent drugs were administered during the latter part (clinical versus subclinical). Of additional importance
of combined treatment. Regardless of the underlying is that with clear pattern of failure, we would be able
principle for such an intervention, these studies nicely to open the door of investigating the determinants of
outlined overall results, relapse-free-survivals and treatment outcome such as cross-resistance between
clearly documented toxicity. The latter was divided drugs or drugs and radiation therapy. This would also
between the concurrent and the consolidation part and lead to investigating more of the inherent nature of
we have all been able to learn more about exact these treatment modalities such as total dose or
toxicity the first or the second part of the treatment fractionation (for radiation therapy) or one or more
were leading to. Unfortunately, this did not happen drug(s) combination (e.g. the same or different drugs
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 423

in the concurrent and the consolidation part of the concurrent cisplatin/etoposide. No difference in
treatment), especially important due to forthcoming toxicity was seen and no statistically significant dif-
generation of drugs waiting to widely enter clinical ference in treatment outcome, although hyperfrac-
practice. tionated radiation therapy offered numerically slightly
Although identifying pattern of failure in patients better survival, and local control.
achieving different response after concurrent radio- Further attempts to optimize the treatment
chemotherapy may require some additional measures approach in this disease include radiation therapy
and likely place additional burden on investigators given concurrently with ‘‘third generation’’ drugs.
and hospitals, this effort would be eventually While all ‘‘third generation’’ drugs have been tested in
rewarding. This way we would be able to discriminate this setting, prospective randomized phase III studies
between different patients and different options and to are lacking. Regardless, it seems that paclitaxel/car-
proceed (or not) with a consolidation therapy in one boplatin combination has similar efficacy and likely
or more patient subsets, an approach which would less toxicity than either cisplatin- or other multiagent-
ultimately lead to better patient-tailored treatment based chemotherapy (Kelly et al. 2001; Schiller et al.
sequence, a must for a clinical research in lung cancer 2002). A number of phase II studies tested this
in the future. combination (Choy et al. 1998; Choy et al. 2000; Lau
One of the unsolved question on ‘‘optimization’’ of et al. 2001; Jeremic et al. 2005) with promising
concurrent radiochemotherapy, particularly from the results. The first prospective study comparing radia-
standpoint of radiation oncology, is the type of frac- tion therapy/paclitaxel versus radiation therapy alone
tionation; conventional, once daily or altered frac- showed advantage for radiation therapy/paclitaxel
tionation, employing multiple fractions per day (the median survival time, 15.2 vs. 12 months;
(hyperfractionation). The Radiation Therapy Oncol- p = 0.027) (Ulutin and Pak 2003). Testing paclitaxel/
ogy Group 8311 study (Cox et al. 1990) showed a carboplatin combination and standard-fraction radia-
possible advantage only for a hyperfractionated tion therapy (63 Gy) in three schedules, Choy et al.
radiation therapy dose of 69.6 Gy, 1.2 Gy b.i.d. (2002) used either pre-radiation therapy chemother-
fractionation (but not beyond it) over standard 60 Gy apy followed by radiation therapy (arm 1), pre-radi-
given in 30 daily fractions in a favourable subset of ation therapy and concurrent radiochemotherapy (arm
locally advanced nonsmall cell lung cancer, Radiation 2), and concurrent radiochemotherapy and post-radi-
Therapy Oncology Group 9410, while not statistically ation therapy chemotherapy (arm 3). Although this
designed to directly compare standard vs. altered phase II randomized study was not designed to sta-
fractionation, appeared to show no survival difference tistically compare treatment arms, nevertheless, the
between conventional, once daily and hyperfraction- best results were achieved in the arm 3 (the median
ated radiation therapy when both given with concur- survival time, 16.1 months; 2-year survival, 33%).
rent chemotherapy. Interestingly, when compared to Also, in the arm 2 there was suboptimal compliance
conventionally fractionated radiation therapy, hyper- with concurrent radiochemotherapy after induction
fractionated radiochemotherapy offered better local chemotherapy in arm 2.
control in the Radiation Therapy Oncology Group
9410, but this did not translate into a difference in
survival. Another study came to the same conclusion, 7 New Approaches in Radiation
albeit of somewhat different treatment approach. In Therapy and Chemotherapy
the North Central Cancer Treatment Group/Mayo of Locally Advanced Nonsmall-Cell
Clinic phase III study (Schild et al. 2002) conven- Lung Cancer
tionally fractionated radiation therapy (60 Gy) was
compared to split-course hyperfractionated radiation Some of the newer approaches regarding the chemo-
therapy using 30 Gy given in 20 fractions in ten therapy have been mentioned above. It is also
treatment days in 2 weeks with a 2-week break after expected that more new drugs will become more
which another 30 Gy were given using the same readily available in the future and that the process of
fractionation. Both conventionally fractionated and their initial clinical testing (phase I–III) include test-
hyperfractionated radiation therapy groups received ing for their radioenhancing potentials which would
424 B. Jeremić et al.

go in parallel to its testing for anticancer chemo- Fukumoto et al. 2002; Nagata et al. 2002; Whyte et al.
therapy purposes. This way, we would be able earlier 2003; Hof et al. 2003; Timmerman et al. 2003;
to learn about drug properties, both alone or in Onimaru et al. 2003) tumours, its application is
combination with radiation therapy and to address slowly extending to tumours being classified as
important issues of optimal sequencing radiation locally advanced. It is not unrealistic to expect that
therapy and chemotherapy in locally advanced extracranial stereotactic radiosurgery and stereotactic
disease. fractionated radiation therapy will play an important
Regarding radiation therapy, wide application of role in metastatic nonsmall cell lung cancer, par-
powerful computers made a substantial impact on ticularly in cases with favourable characteristics
treatment planning and delivery. Three-dimensional (response to chemotherapy, single metastatic lesion,
conformal radiation therapy is now increasingly being small primary tumours, etc.). However, it should be
practiced worldwide. With radiation therapy fields clearly emphasized that proper selection of patients
tailored to include only detectable tumour, more remains prerequisite for the use of these new tech-
focused and escalated radiation therapy doses can be nologies in locally advanced and/or metastatic non-
given. Phase I/II studies have shown that radiation small cell lung cancer. On the other side, proper
therapy doses at the order of C80 Gy are being fre- selection of patients suitable for any form of com-
quently used with acceptable toxicity (Armstrong bined radiation therapy and chemotherapy, may
et al. 1993, 1997; Robertson et al. 1997), and that the enable using the two modality approach in non-
radiation therapy concepts of the necessity of elective curative approach. In such one study, Nawrocki et al.
nodal irradiation may be challenged. It should, how- (2010) reported on a of a randomized phase II study
ever, be mentioned that with even with limited field using (A) palliative radiation therapy given alone
radiation therapy in three-dimensional conformal (30 Gy/10 fractions) versus (B) the same palliative
radiation therapy, some incidental elective nodal radiation therapy given with concurrent chemother-
irradiation always occur, and may approach apy (two cycles of cisplatin and vinorelbine followed
45–50 Gy, considered as the radiation therapy dose by radiation therapy together with third cycle) in
necessary for elective treatment (Martel et al. 1999; patients with stage IIIA to IIIB nonsmall cell lung
Rosenzweig et al. 2001). In addition to increasing cancer not eligible for radical (tumor [8 cm and/or
target coverage and allowing radiation dose escala- forced expiratory volume B40%, performance status
tion, the use of three-dimensional conformal radiation 0–2, and existing tumour-related chest symptoms) A
therapy also allows more accurate prediction of tox- total of 99 patients were eligible for response, overall
icity of a given course of radiation therapy (Graham survival, and progression-free survival evaluation.
1997; Kwa et al. 1998). Median age was 66 years (45–78 years). Response
Intensity-modulated radiotherapy has also been rate was 27% vs. 53%, p = 0.08; median overall
used in locally advanced nonsmall cell lung cancer survival was 9.0 vs. 12.9 months, p = 0.0342; and
and potential advantages of intensity-modulated median progression-free survival was 4.7 vs.
radiation therapy became evident when one compares 7.3 months, p = 0.046, in arm A versus arm B,
the three-dimensional conformal radiation therapy respectively. There were no deaths during treatment
and intensity-modulated radiation therapy plans in arm A and six deaths in arm B; no haematological
(Yorke 2001). With intensity-modulated radiation grade 3–4 toxicities in arm A and 14 toxicities in arm
therapy, in majority of cases the prescription dose B. Symptom control was high and similar in both
could be increased. This was coupled with the arms. Upfront chemotherapy combined with palliative
decreased lung dose and improved planning target radiation therapy (30 Gy) may be a promising treat-
volume uniformity, as well as significantly reducing ment option in the subpopulation of patients with
cumulative radiation therapy dose to the oesophagus stage IIIA to IIIB non-small cell lung cancer not
while maintaining the same or higher dose to gross amenable for definitive radiochemotherapy and
disease (Giraud et al. 2001). While extracranial ste- deserves further investigation.
reotactic radiosurgery and stereotactic fractionated Although hardly termed as ‘‘new approach’’, the
radiation therapy were initially used only for small use of radioprotectors faced renewed clinical interest
(early stage) (Uematsu et al. 1998; Hara et al. 2002; in a protection of radiation therapy-induced toxicity.
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 425

Several studies reported on the use of amifostine pathologically staged as IIIB and a minority of those
during radiation therapy and chemotherapy in lung staged as IIIA will actually have metastatic burden
cancer. Antonodou et al. (2001) performed a ran- from the outset, undiagnosed with current diagnostic
domized phase III trial of radiation therapy with or tools. This is supported by simple observation of the
without daily amifostine in patients with advanced natural history of this disease, regardless of the
stage lung cancer. The incidence of pneumonitis C2 treatment: there are always some patients who fail
was significantly lower in the amifostine group as distantly several months or a year after the diagnosis.
well as incidence of esophagitis Cgrade 2, and the These patients are likely to be ones who would be
protective effect of amifostine enabled lower inci- upstaged by the use of positron emission tomography
dence of late damage, too, with no effect on treatment and moved to stage IV (metastatic disease). On the
outcome. Further evidence came from Komaki et al. other hand, it is expected that a proportion of patients
(2002) who administered amifostine twice weekly with early stages (I and II) nonsmall cell lung cancer
before treatment in patients with inoperable nonsmall will also be upstaged, and will likely increase the
cell lung cancer treated with concurrent radioche- number of patients actually having stage III (locally
motherapy. They observed that Morphine intake to advanced) disease. Whatever predominates, locally
reduce severe esophagitis was significantly lower in advanced nonsmall cell lung cancer will remain one
the amifostine arm as well as was the incidence of of the major focuses of clinical research in lung
acute pneumonitis in the treatment arm. Finally, a cancer simply because major improvements occurred
randomized double-blind study (Leong et al. 2003) here and they have occurred owing to optimized
showed a trend for fewer patients showing toxicity in combined modality treatments, notably combined
the amifostine group. The Radiation Therapy Oncol- radiation therapy and chemotherapy. There is con-
ogy Group has just reported preliminary results on tinued discussion regarding the role of surgery for
study Radiation Therapy Oncology Group 98-01, these patients (Taylor et al. 2004). Four randomized
which randomized patients to intensive chemoradia- studies note no overall survival differences comparing
tion (Induction carboplatin/paclitaxel followed by operative vs. non-operative approaches in patients
hyperfractionated radiation therapy to 69.6 Gy with with stage III lung cancer (Shepherd et al. 1998;
concurrent weekly carboplatin/paclitaxel) with or Johnstone et al. 2002; van Meerbeeck et al. 2007;
without amifostine four times per week during radi- Albain et al. 2009). An unplanned subset analysis of
ation therapy. Although there was no difference in the the most contemporary of these trials, Intergroup-
rate of Grade 3 esophagitis, patient-reported area- 0139 (Albain et al. 2009), did suggest a difference on
under-the-curve swallowing dysfunction scores were survival based on surgical approach. That is, mortality
significantly lower in the amifostine group (Movsas rates with pneumonectomy were excessively high,
et al. 2003) It is expected that more studies regarding while lobectomy patients appeared to have improved
the issue of optimal protection with amifostine will outcomes. It remains nonetheless appropriate to con-
provide more data on further optimization before clude that the sum of the evidence to date supports the
becoming a standard adjunct to radiation therapy or proposition that a non-surgical approach constitutes
radiochemotherapy treatments in the future. the ‘‘standard’’ for stage III patients. Radiation ther-
apy and chemotherapy will therefore, further evolve
in the near future and will bring us to the exiting era
8 Conclusions of more successful clinical research, leading ulti-
mately to better outcome in this disease.
Locally advanced nonsmall cell lung cancer is one of
the major targets for clinical research in lung cancer.
While it was accounted for approximately 40% of all
References
cases, it is expected that widespread use of positron
emission tomography (leading to more precise staging
Akerley W, Herndon JE Jr, Lyss AP, Choy H, Turrisi A,
of patients) will likely decrease in the number of Graziano S, Williams T, Zhang C, Vokes EE, Green MR
patients falling into stage III nonsmall cell lung can- (2005) Induction paclitaxel/carboplatin followed by con-
cer. This is because patients clinically or even current chemoradiation therapy for unresectable stage III
426 B. Jeremić et al.

non-small-cell lung cancer: a limited-access study—CAL- for patients with unresectable stage IIIA and B non-small-cell
GB 9534. Clin Lung Cancer 7:47–53 lung cancer. J Clin Oncol 23:3760–3767
Albain KS, Crowley JJ, Turrisi AT III, Gandara DR, Farrar Belderbos J, Uitterhoeve L, van Zandwijk N, Belderbos H,
WB, Clark JI, Beasley KR, Livingston RB (2002) Concur- Rodrigus P, van de Vaart P, Price A, van Walree N, Legrand
rent cisplatin, etoposide, and chest radiotherapy in patho- C, Dussenne S, Bartelink H, Giaccone G, Koning C,
logic stage IIIB non-small-cell lung cancer: a Southwest EORTC LCG and RT Group (2007) Randomised trial of
Oncology Group Phase II study, SWOG 9019. J Clin Oncol sequential versus concurrent chemo-radiotherapy in patients
20:3454–3460 with inoperable non-small cell lung cancer (EORTC 08972-
Albain KS, Swann RS, Rusch VW, Turrisi AT 3rd, Shepherd 22973). Eur J Cancer 43:114–121
FA, Smith C, Chen Y, Livingston RB, Feins RH, Gandara Blanke C, Ansari R, Mantravadi R, Gonin R, Tokars R, Fisher
DR, Fry WA, Darling G, Johnson DH, Green MR, Miller W, Pennington K, O’Connor T, Rynard S, Miller M (1995)
RC, Ley J, Sause WT, Cox JD (2009) Radiotherapy plus Phase III trial of thoracic irradiation with or without
chemotherapy with or without surgical resection for stage cisplatin for locally advanced unresectable non-small cell
III non-small-cell lung cancer: a phase III randomised lung cancer: a Hoosier Oncology Group Protocol. J Clin
controlled trial. Lancet 374:379–386 Oncol 13:1425–1429
Antonodou D, Coliarakis N, Synodinou M, Athanassiou H, Bonner JA, McGinnis WL, Stella PJ, Marschke RF Jr, Sloan
Kouveli A, Verigos C, Georgakopolous G, Panousaaki K, JA, Shaw EG, Mailliard JA, Creagan ET, Ahuja RK,
Karageorgis P, Throuvalas N, Clinical Radiation Oncology Johnson PA (1998) The possible advantage of hyperfrac-
Hellenic Group (2001) Randomized phase III trial of tionated thoracic radiotherapy in the treatment of locally
radiation treatment +/- amifostine in patients with advanced non small cell lung cancer. Results of a North
advanced-stage lung cancer. Int J Radiat Oncol Biol Phys Central Cancer Treatment Group phase III study. Cancer
51:915–922 82:1037–1048
Armstrong JG, Burman C, Leibel SA, Fontenla D, Kutcher G, Bozcuk H, Artac M, Ozdogan M (2010) Correlates of benefit
Zelefsky M, Fuks Z (1993) Three-dimensional conformal from neoadjuvant chemotherapy before radiotherapy in non-
radiation therapy may improve the therapeutic ratio of high small cell lung cancer: a meta-analytical approach with
dose radiation therapy for lung cancer. Int J Radiat Oncol meta-regression analysis. J BUON 15:43–50
Biol Phys 26:685–689 Byhardt RW (1999) Toxicities in RTOG combined-modality
Armstrong JG, Raben A, Zelefsky M, Burt M, Leibel SA, trials for inoperable non-small-cell lung cancer. Oncology
Burman C, Kutcher G, Harrison LB, Hahn C, Ginsberg R, (Huntingt) 13(10 Suppl 5):116–120
Rusch V, Kris M, Fuks Z (1997) Promising survival with Byhardt RW, Pajak TF, Emami B, Herskovic A, Doggett RS,
three-dimensional conformal radiation therapy for non- Olsen LA (1993) A phase I/II study to evaluate accelerated
small cell lung cancer. Radiother Oncol 44:17–22 fractionation via concomitant boost for squamous, adeno,
Arriagada R, Le Chevalier T, Rekacewicz C, Quoix E, De and large cell carcinoma of the lung: report of Radiation
Cremoux H, Douillard JY, Tarayre M (1997) Cisplatin- Therapy Oncology Group 84-07. Int J Radiat Oncol Biol
based chemotherapy (CT) in patients with locally advanced Phys 26:459–468
non-small cell lung cancer (NSCLC): late analysis of a Byhardt RW, Scott CB, Ettinger DS, Curran WJ, Doggett RL,
French randomized trial. Proc Am Soc Clin Oncol 16:16 Coughlin C, Scarantino C, Rotman M, Emami B (1995)
(abstract) Concurrent hyperfractionated irradiation and chemotherapy
Aupérin A, Le Péchoux C, Rolland E, Curran WJ, Furuse K, for unresectable nonsmall cell lung cancer. Results of
Fournel P, Belderbos J, Clamon G, Ulutin HC, Paulus R, Radiation Therapy Oncology Group 90-15. Cancer 75:
Yamanaka T, Bozonnat MC, Uitterhoeve A, Wang X, 2337–2344
Stewart L, Arriagada R, Burdett S, Pignon JP (2010) Meta- Byhardt RW, Scott C, Sause WT, Emami B, Komaki R, Fisher
analysis of concomitant versus sequential radiochemother- B, Lee JS, Lawton C (1998) Response, toxicity, failure
apy in locally advanced non-small-cell lung cancer. J Clin patterns, and survival in five Radiation Therapy Oncology
Oncol 28:2181–2190 Group (RTOG) trials of sequential and/or concurrent
Ball D, Bishop J, Smith J, O’Brien P, Davis S, Ryan G, Olver I, chemotherapy and radiotherapy for locally advanced non-
Toner G, Walker Q, Joseph D (1999) A randomised phase small-cell carcinoma of the lung. Int J Radiat Oncol Biol
III study of accelerated or standard fraction radiotherapy Phys 42:469–478
with or without concurrent carboplatin in inoperable non- Cakir S, Egehan I (2004) A randomized clinical trial of
small cell lung cancer: final report of a multi-centre trial. radiotherapy plus cisplatin versus radiotherapy alone in
Radiother Oncol 52:129–136 stage III non-small cell lung cancer. Lung Cancer 43:309–
Baumann M, Hermann T, Koch R (2005) Continuous 316
hyperfractionated accelerated radiotherapy weekend-less Choy H, Akerley W, Safran H, Graziano S, Chung C, Williams
versus conventionalloy fractionated (CF) in non-small cell T, Cole B, Kennedy T (1998) Multiinstitutional phase II
lung cancer (NSCLC): first results of a phase III random- trial of paclitaxel, carboplatin, and concurrent radiation
zied multicentre trial (ARO 97-91). Eur J Cancer 3(Suppl): therapy for locally advanced non-small-cell lung cancer.
S323 J Clin Oncol 16:3316–3322
Belani CP, Wang W, Johnson DH (2005) Phase III study of the Choy H, DeVore RF, Hande KR, Porter LL, Rosenblatt P,
Eastern Cooperative Oncology Group (ECOG 2597): induc- Yunus F, Schlabach L, Smith C, Shyr Y, Johnson DH
tion chemotherapy followed by either standard thoracic (2000) A phase II study of paclitaxel, carboplatin, and
radiotherapy or hyperfractionated accelerated radiotherapy hyperfractionated radiation therapy for locally advanced
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 427

inopearble non-small cell lung cancer (a Vanderbilt cancer El Sharouni SY, Kal HB, Battermann JJ (2003) Accelerated
center affiliate network study). Int J Radiat Oncol Biol Phys regrowth of non-small-cell lung tumors after induction
47:931–937 chemotherapy. Br J Cancer 89:2184–2189
Choy H, Curran WJ, Scott CB (2002) Preliminary report of Fournel P, Robinet G, Thomas P, Souquet PJ, Léna H,
locally advanced multimodality protocol (LAMP): ACR Vergnenégre A, Delhoume JY, Le Treut J, Silvani JA,
427: a randomized phase II study of three chemo-radiation Dansin E, Bozonnat MC, Daurés JP, Mornex F, Pérol M,
regimens with paclitaxel, carboplatin, and thoracic radiation Groupe Lyon-Saint-Etienne d’Oncologie Thoracique-
(TRT) for patients with locally advanced non small cell lung Groupe Français de Pneumo-Cancérologie (2005) Random-
cancer (LA-NSCLC). Proc Am Soc Clin Oncol, pp 291a ized phase III trial of sequential chemoradiotherapy com-
(abstr. #1160) pared with concurrent chemoradiotherapy in locally
Clamon G, Herndon J, Cooper R, Chang AY, Rosenman J, advanced non-small-cell lung cancer: Groupe Lyon-Saint-
Green MR (1999) Radiosensitization with carboplatin for Etienne d’Oncologie Thoracique-Groupe Français de
patients with unresectable stage III non-small-cell lung Pneumo-Cancérologie NPC 95-01 study. J Clin Oncol
cancer: a phase III trial of the cancer and Leukemia group B 23:5910–5917
and the Eastern Cooperative Oncology Group. J Clin Oncol Fukumoto S, Shirato H, Shimizu S, Ogura S, Onimaru R,
17:4–11 Kitamura K, Yamazaki K, Miyasaka K, Nishimura M,
Cox JD, Yesner R, Mietlowski W, Petrovich Z (1979) Influence Dosaka-Akita H (2002) Small-volume image-guided radio-
of cell type on failure pattern after irradiation for locally therapy using hypofractionated coplanar and noncoplanar
advanced carcinoma of the lung. Cancer 44:94–98 multiple fields with inoperable stage I nonsmall cell lung
Cox JD, Azarnia N, Byhardt RW, Shin KH, Emami B, Pajak TF carcinomas. Cancer 95:1546–1553
(1990) A randomized phase I/II trial of hyperfractionated Fukuoka M, Niitani H, Suzuki A, Motomiya M, Hasegawa K,
radiation therapy with total doses of 60.0 Gy to 79.2 Gy: Nishiwaki Y, Kuriyama T, Ariyoshi Y, Negoro S, Masuda
possible survival benefit with [69.6 Gy in favorable N (1992) Phase II study of CPT-11, a new derivative of
patients with Radiation Therapy Oncology Group stage III camptothecin for previously untreated non-small cell lung
non-small cell lung carcinoma: report of Radiation Therapy cancer. J Clin Oncol 10:16–20
Oncology Group 83-11. J Clin Oncol 8:1543–1555 Furuse K, Nishikawa H, Takada Y, Nishikawa H, Takada Y,
Cox JD, Pajak TF, Asbell S, Russell AH, Pederson J, Byhardt Kudoh S, Katagami N, Ariyoshi Y (1999) Phase III study of
RW, Emami B, Roach M 3rd (1993) Interruptions of high- concurrent versus sequential thoracic radiotherapy in com-
dose radiation therapy decrease long-term survival of bination with mitomycin, vindesine and cisplatin in unre-
favorable patients with unresectable non-small cell carci- sectable stage III non-small-cell lung cancer. J Clin Oncol
noma of the lung: analysis of 1244 cases from 3 Radiation 17:2692–2699
Therapy Oncology Group (RTOG) trials. Int J Radiat Oncol Furuse K, Hosoe S, Masuda N (2000) Impact of tumor control
Biol Phys 27:493–498 on survival in unresectable stage III non-small cell lung
Crino L, Latini P, Meacci M, Corgna E, Maranzano E, Darwish cancer (NSCLC) treated with concurrent thoracic radiother-
S, Minotti V, Santucci A, Tonato M (1993) Induction apy (TRT) and chemotherapy (CT). Proc Am Soc Clin
chemotherapy plus high-dose radiotherapy versus radio- Oncol 19 (Abstract 1893)
therapy alone in locally advanced unresectable non-small- Gandara DR, Chansky K, Albain KS, Leigh BR, Gaspar LE,
cell lung cancer. Ann Oncol 4:847–851 Lara PN Jr, Burris H, Gumerlock P, Kuebler JP, Bearden JD
Cullen MH, Billingham LJ, Woodroffe CM, Gower N, Souhami 3rd, Crowley J, Livingston R (2003) Consolidation doce-
RL, Chetiyawaedana AD, Joshi R, Rudd R, Trask C, Spiro S taxel after concurrent chemoradiotherapy in stage IIIB non-
(1997) Mitomycin, ifosfamide and cisplatin (MIC) in non- small-cell lung cancer: phase II Southwest Oncology Group
small cell lung cancer (NSCLC): 1. Results of a randomised Study S9504. J Clin Oncol 21:2004–2010
trial in patients with localised, inoperable disease. Lung Giraud P, Rosenzweig KE, Yorke E (2001) Radiotherapy for
Cancer 18(Suppl 1):5 (abstract 10) lung cancer: can IMRT decrease the risk of esophagitis?
Curran WJ Jr, Scott C, Langer C (2000) Phase III comparison Proc Am Soc Ther Radiol Oncol (ASTRO). San Francisco,
of sequential Vs concurrent chemoradiation for pts with Int J Radiat Oncol Biol Phys, pp 355–356 (abstr. #2250)
unresected stage III non-small cell lung cancer (NSCLC): Goldstraw P, Crowley JJ (2006) The International Association
initial report of Radiation Therapy Oncology Group for the Study of Lung Cancer International staging project
(RTOG) 9410. Proc Am Soc Clin Oncol 19:484a (Abstract on lung cancer. J Thorac Oncol 1:281–286
1891) Goldstraw P, Crowley J, Chansky K, Giroux DJ, Groome PA
Dillman RO, Seagren SL, Propert KJ, Guerra J, Eaton WL, (2007) The IASLC lung cancer staging project: proposals
Perry MC, Carey RW, Frei EF 3rd, Green MR (1990) A for the revision of the TNM stage groupings in the
randomized trial of induction chemotherapy plus high-dose forthcoming (seventh) edition of the TNM classification of
radiation versus radiation alone in stage III non-small-cell malignant tumours. J Thorac Oncol 2:706–714
lung cancer. N Engl J Med 323:940–945 Graham MV (1997) Predicting radiation response. Int J Radiat
Dillman RO, Herndon J, Seagren SL, Eaton WL Jr, Green MR Oncol Biol Phys 39:561–562
(1996) Improved survival in stage III non-small-cell lung Groen HJG, van de Leest AHW, Fokkema E, Timmer PR,
cancer: seven-year follow-up of Cancer and Leukemia Nossent GD, Smit WJ, Nabers J, Oosterhuis B, Hoekstra HJ,
Group B (CALGB) 8433 trial. J Natl Cancer Inst 88:1210– Hermans J, Otter R, van Putten JWG, De Vries EG, Mulder
1215 NH (2004) Phase III study of continuous carboplatin over
428 B. Jeremić et al.

6 weeks with radiation versus radiation alone in stage III daily carboplatin/paclitaxel in patients with stage III non-
non small cell lung cancer. Ann Oncol 15:427–432 small cell lung cancer (NSCLC). Long-term results of a
Groome PA, Bolejack V, Crowley JJ, Kennedy C, Krasnik M phase II study. J Clin Oncol 23:1144–1151
(2007) The IASLC lung cancer staging project: validation Johnson DH, Einhorn LH, Bartolucci A, Birch R, Omura G,
of the proposals for revision of the T, N, and M descriptors Perez CA, Greco FA (1990) Thoracic radiotherapy does not
and consequent stage grouping in the forthcoming (seventh) prolong survival in patients with locally advanced unresec-
edition of TNM classification of malignant tumours. table non-small cell lung cancer. Ann Int Med 113:33–38
J Thorac Oncol 2:694–705 Johnson DH, Paul DM, Hande KR, Shyr Y, Blanke C, Murphy
Hanna N, Neubauer M, Yiannoutsos C, McGarry R, Arseneau J, B, Lewis M, De Vore RF 3rd (1996) Paclitaxel plus
Ansari R, Reynolds C, Govindan R, Melnyk A, Fisher W, carboplatin in advanced non-small-cell lung cancer: a phase
Richards D, Bruetman D, Anderson T, Chowhan N, Nattam S, II trial. J Clin Oncol 14:2054–2060
Mantravadi P, Johnson C, Breen T, White A, Einhorn L, Johnstone DW, Byhardt RW, Ettinger D, Scott CB (2002)
Hoosier Oncology Group; US Oncology (2008) Phase III study Phase III study comparing chemotherapy and radiotherapy
of cisplatin, etoposide, and concurrent chest radiation with or with preoperative chemotherapy and surgical resection in
without consolidation docetaxel in patients with inoperable patients with non-small-cell lung cancer with spread to
stage III non-small-cell lung cancer: the Hoosier Oncology mediastinal lymph nodes (N2); final report of RTOG 89-01.
Group and U.S. Oncology. J Clin Oncol 26:5755–5760 Radiation Therapy Oncology Group. Int J Radiat Oncol Biol
Hara R, Itami J, Kondo T, Aruga T, Abe Y, Ito M, Fuse M, Phys 54:365–369
Shinohara D, Nagaoka T, Kobiki T (2002) Stereotactic Kelly K, Crowley J, Bunn PA Jr, Presant CA, Grevstad PK,
single high dose irradiation of lung tumors under respiratory Moinpour CM, Ramsey SD, Wozniak AJ, Weiss GR, Moore
gating. Radiother Oncol 63:159–163 DF, Israel VK, Livingston RB, Gandara DR (2001)
Harada H, Yamamoto N, Takahashi T, Endo M, Murakami H, Randomized phase III trial of paclitaxel plus carboplatin
Tsuya A, Nakamura Y, Ono A, Igawa S, Shukuya T, versus vinorelbine plus cisplatin in the treatment of patients
Tamiya A, Nishimura T (2009) Comparison of chemother- with advanced NSCLC: a SWOG trial. J Clin Oncol
apy regimens for concurrent chemoradiotherapy in unre- 19:3210–3218
sectable stage III non-small cell lung cancer. Int J Clin Kelly K, Chansky K, Gaspar LE, Albain KS, Jett J, Ung YC,
Oncol. 14:507–512 Lau DH, Crowley JJ, Gandara DR (2008) Phase III trial of
Herscher LL, Hahn SM, Kroog G, Pass H, Temeck B, Goldspiel maintenance gefitinib or placebo after concurrent chemora-
B, Cook J, Mitchell JB, Liebmann J (1998) Phase I study of diotherapy and docetaxel consolidation in inoperable stage
paclitaxel as a radiation sensitizer in the treatment of III non-small-cell lung cancer: SWOG S0023. J Clin Oncol
mesothelioma and non-small-cell lung cancer. J Clin Oncol 26:2450–2456
16:635–641 Komaki R, Scott C, Ettinger D, Lee JS, Fossella FV, Curran
Hof H, Herfarth K, Munter M, Hoess A, Motsch J, Wannenm- W, Evans RF, Rubin P, Byhardt RW (1997) Randomized
acher M, Debus J (2003) Stereotactic single-dose radiother- study of chemotherapy/radiation therapy combinations for
apy of stage I non-small-cell lung cancer (NSCLC). Int J favorable patients with locally advanced inoperable non-
Radiat Oncol Biol Phys 56:335–341 small cell lung cancer: Radiation Therapy Oncology
Holsti LR, Matson K (1980) A randomized study of split-course Group (RTOG) 92-04. Int J Radiat Oncol Biol Phys 38:
radiotherapy of lung cancer: long term results. Int J Radiat 149–155
Oncol Biol Phys 6:977–981 Komaki R, Seiferheld W, Curran Wl (2000) Sequential vs.
Jeremic B, Shibamoto Y, Acimovic L, Djuric L (1995) concurrent chemotherapy, radiation therapy for inoperable
Randomized trial of hyperfractionated radiation therapy non-small cell lung cancer (NSCLC): analysis of failures in
with or without concurrent chemotherapy for stage III non- a phase III study (RTOG 9410). Proc Am Soc Ther Radiol
small-cell lung cancer. J Clin Oncol 13:452–458 Oncol 42:113 (Abstract 5)
Jeremic B, Shibamoto Y, Acimovic Lj, Milisavljevic S (1996) Komaki R, Lee JS, Kaplan B, Allen P, Kelly JF, Liao Z,
Hyperfractionated radiation therapy with or without con- Stevens CW, Fossella FV, Zinner R, Papadimitrakopolou V,
current low-dose daily carboplatin/etoposide for stage III Khuri F, Glisson B, Pisters K, Kurie J, Herbst R, Milas L,
non-small-cell lung cancer: a randomized study. J Clin Ro J, Thames HD, Hong WK, Cox JD (2002) Randomized
Oncol 14:1065–1070 phase III study of chemoradiation with or without amifos-
Jeremic B, Shibamoto Y, Milicic B, Nikolic N, Dagovic A, tine for patients with favorable performance status inoper-
Milisavljevic S (1998) Concurrent radiochemotherapy for able stage II–III non-small cell lung cancer: preliminary
patients with stage III non-small cell lung cancer (NSCLC). results. Semin Radiat Oncol 12(1 Suppl. 1):46–49
Long-term results of a phase II study. Int J Radiat Oncol Kwa Sl, Lebesque JV, Theuws JC, Marks LB, Munley MT,
Biol Phys 42:1091–1096 Bentel G, Oetzel D, Spahn U, Graham MV, Drzymala RE,
Jeremic B, Shibamoto Y, Acimovic LJ et al (2001) Hyperfrac- Purdy JA, Lichter AS, Martel MK, Ten Haken RK (1998)
tionated radiation therapy and concurrent low-dose, daily Radiation pneumonitis as a function of mean lung dose: an
carboplatin/etoposide with or without week-end carboplatin/ analysis of pooled data of 540 patients. Int J Radiat Oncol
etoposide chemotherapy in stage III non-small-cell lung Biol Phys 42:1–9
cancer: a randomized trial. Int J Radiat Oncol Biol Phys Lau D, Leigh B, Gandara D, Edelman M, Morgan R, Israel V,
50:19–25 Lara P, Wilder R, Ruy J, Doroshow J (2001) Twice-weekly
Jeremic B, Milicic B, Acimovic L, Milisavljevic S (2005) paclitaxel and weekly carboplatin with concurrent thoracic
Concurrent hyperfractionated radiotherapy and low-dose radiation followed by carboplatin/paclitaxel consolidation
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 429

for stage III non-small-cell lung cancer: a California cancer Nordman E, Platin L-H, Pyrhonen S, Romppanen M-L,
consortium phase II study. J Clin Oncol 19:442–447 Salmi R, Tammilehto L, Taskinen PJ (1988) Inoperable
Le Chevalier T, Arriagada R, Quoix E, Ruffie P, Martin M, non-small cell lung cancer: radiation with or without
Tarayre M, Lacombe-Terrier MJ, Douillard JY, Laplanche chemotherapy. Eur J Cancer Clin Oncol 24:477–482
A (1991) Radiotherapy alone versus combined chemother- Mauer AM, Masters GA, Haraf DJ, Hoffman PC, Watson SM,
apy and radiotherapy in nonresectable non-small-cell lung Golomb HM, Vokes EE (1998) Phase I study of docetaxel
cancer: first analysis of a randomized trial in 353 patients. with concomitant thoracic radiation therapy. J Clin Oncol
J Natl Cancer Inst 83:417–423 16:159–164
Le Chevalier T, Arriagada R, Tarayre M, Lacombe-Terrier MJ, Mehta MP, Tannehill SP, Adak S, Martin L, Petereit DG,
Laplanche A, Quoix E, Ruffie P, Martin M, Douillard JY Wagner H, Fowler JF, Johnson D (1998) Phase II trial of
(1992) Significant effect of adjuvant chemotherapy on hyperfractionated accelerated radiation therapy for nonre-
survival in locally advanced non-small cell lung carcinoma. sectable non-small-cell lung cancer: results of Eastern
J Natl Cancer Inst 84:58 (letter) Cooperative Oncology Group 4593. J Clin Oncol 16:
Le Chevalier T, Brisgand D, Douillard J-Y, Pujol JL, Alberola 3518–3523
V, Monnier A, Riviere A, Lianes P, Chomy P, Cigolari S Millward MJ, Zalcberg J, Bishop JF, Webster LK, Zimet A,
(1994) Randomized study of vinorelbine and cisplatin Rischin D, Toner GC, Laird J, Cosolo W, Urch M, Bruno R,
versus vindesine and cisplatin versus vinorelbine alone in Loret C, James R, Blanc C (1996) Phase I trial of docetaxel
advanced non-small-cell lung cancer: results of a European and cisplatin in previously untreated patients with advanced
multicenter trial including 612 patients. J Clin Oncol non-small cell lung cancer. J Clin Oncol 14:750–758
12:360–367 Morton RF, Jett JR, McGinnis WL, Earle JD, Therneau TM,
Lee JS, Scott C, Komaki R, Fossella FV, Dundas GS, Krook JE, Elliott TE, Mailliard JA, Nelimark RA, Mak-
McDonald S, Byhardt RW, Curran WJ Jr (1996) Concurrent symiuk AW, Drummond RG, Laurie JA, Kugler JW,
chemoradiation therapy with oral etoposide and cisplatin for Anderson RT (1991) Thoracic radiation therapy alone
locally advanced inoperable non-small-cell lung cancer: compared with combined chemoradiotherapy for locally
Radiation Therapy Oncology Group protocol 91-06. J Clin unresectable non-small cell lung cancer. A randomized,
Oncol 14:1055–1064 phase III trial. Ann Intern Med 115:681–686
Leong SS, Tan EH, Fong KW, Wilder-Smith E, Ong YK, Tai Mountain CF (1986) A new international staging system for
BC, Chew L, Liem SH, Wee J, Foo KM, Ang P, Ang PT lung cancer. Chest 89:225S–233S
(2003) Randomized double-blind trial of combined modality Mountain CF (1997) Revisions in the international system for
treatment with or without amifostine in unresectable stage III staging lung cancer. Chest 111:1710–1717
non-small-cell lung cancer. J Clin Oncol 21:1767–1774 Movsas B, Scott C, Langer C (2003) Phase III study of
Liang HY, Zhou H, Li XL, Yin ZH, Guan P, Zhou BS (2010) amifostine in patients with locally advanced non-small cell
Chemo-radiotherapy for advanced non-small cell lung lung cancer (NSCLC) receiving chemotherapy & hyper-
cancer: concurrent or sequential? It’s no longer the ques- fractionated radiation (chemo/HFxRT): Radiation Therapy
tion: a systematic review. Int J Cancer 127:718–728 Oncology Group (RTOG) 98-01. Proc Am Soc Clin Oncol
Lynch TJ Jr, Kalish L, Strauss G, Elias A, Skarin A, Shulman 21 : 636 (Abstract 2559)
LN, Posner M, Frei E 3rd (1994) Phase II study of topotecan Nagata Y, Negoro Y, Aoki T, Mizowaki T, Takayama K,
in metastatic non-small-cell lung cancer. J Clin Oncol Kokubo M, Araki N, Mitsumori M, Sasai K, Shibamoto Y,
12:347–352 Koga S, Yano S, Hiraoka M (2002) Clinical outcomes of 3D
Manegold C, Bergman B, Chemaissani A, Dornoff W, Drings conformal hypofractionated single high-dose radiotherapy
P, Kellokumpu-Lehtinen P, Liippo K, Mattson K, van Pawel for one or two lung tumors using a stereotactic body frame.
J, Ricci S, Sederholm C, Stahel RA, Wagenius G, van Int J Radiat Oncol Biol Phys 52:1041–1046
Walree N, ten Bokkel-Huinink W (1997) Single-agent Nawrocki S, Krzakowski M, Wasilewska-Tesluk E, Kowalski D,
gemcitabine versus cisplatin-etoposide: early results of a Rucinska M, Dziadziuszko R, Sowa A (2010) Concurrent
randomized phase II study in locally-advanced or metastatic chemotherapy and short course radiotherapy in patients with
non-small cell lung cancer. Ann Oncol 8:525–529 stage IIIA to IIIB non-small cell lung cancer not eligible for
Marino P, Preatoni A, Cantoni A (1995) Randomized trials of radical treatment: results of a randomized phase II study.
radiotherapy alone versus combined chemotherapy and J Thorac Oncol 5:1255–1262
radiotherapy in stages IIIa and IIIb nonsmall cell lung Non-small Cell Lung Cancer Collaborative Group (1995)
cancer. A meta-analysis. Cancer 76:593–601 Chemotherapy in non-small cell lung cancer: a meta-
Martel MK, Sahijdak WM, Hayman JA (1999) Incidental dose analysis using updated data on individual patients from 52
to clinically negative nodes from conformal treatment fields randomised clinical trials. Brit Med J 311:899–909
for nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys O’Rourke N, Roqué I, Figuls M, Farré Bernadó N, Macbeth F
45(3S):244 (Abstract) (2010) Concurrent chemoradiotherapy in non-small cell
Masters GA, Haraf DJ, Hoffman PC, Drinkard LC, Krauss SA, lung cancer. Cochrane Database Syst Rev 6:CD002140
Ferguson MK, Olak J, Samuels BL, Golomb HM, Vokes EE Onimaru R, Shirato H, Shimizu S, Kitamura K, Xu B,
(1998) Phase I study of vinorelbine, cisplatin and concom- Fukumoto S, Chang T-C, Fujita K, Oita M, Miyasaka K,
itant thoracic radiation in the treatment of advanced chest Nishimura M, Dosaka-Akita H (2003) Tolerance of organs
malignancies. J Clin Oncol 16:2157–2163 at riskin small-volume, hypofractionated, image-guided
Mattson K, Holsti LR, Holsti P, Jakobsson M, Kajanti M, radiotherapy for primary and metastatic lung cancers. Int J
Liippo K, Mantyla M, Niitamo-Korhonen S, Nikkanen V, Radiat Oncol Biol Phys 56:126–135
430 B. Jeremić et al.

Oshita F, Noda K, Nishiwaki Y, Fujita A, Kurita Y, Nakabay- radiation therapy. Int J Radiat Oncol Biol Phys 37:1079–
ashi T, Tobise K, Abe S, Suzuki S, Hayashi I, Kawakami Y, 1085
Matsuda T, Tsuchiya S, Takahashi S, Tamura T, Saijo N Rosenzweig KE, Sim SE, Mychalczak B, Braban LE, Schindel-
(1997) Phase II study of irinotecan and etoposide in patients heim R, Leibel SA (2001) Elective nodal irradiation in the
with metastatic non-small-cell lung cancer. J Clin Oncol treatment of non-small-cell lung cancer with three-dimen-
15:304–309 sional conformal radiation therapy. Int J Radiat Oncol Biol
Perez CA, Bauer M, Edelstein S, Gillespie BW, Birch R (1986) Phys 50:681–685
Impact of tumor control on survival in carcinoma of the Roswit B, Patno ME, Rapp R, Veinbergs A, Feder B, Stuhlbarg
lung treated with irradiation. Int J Radiat Oncol Biol Phys J, Reid CB (1968) The survival of patients with inoperable
12:539–547 lung cancer: a large-scale randomized study of radiation
Perez CA, Pajak TF, Rubin P, Simpson JR, Mohiuddin M, therapy versus placebo. Radiology 90:688–697
Brady LW, Perez-Tamayo R, Rotman M (1987) Long term Rusch VW, Crowley J, Giroux DJ, Goldstraw P, Im JG (2007) The
observations of the patterns of failure in patients with IASLC lung staging project: proposals for the revision of the N
unresectable non-oat cell carcinoma of the lung treated with descriptors in the forthcoming (seventh) edition of the TNM
definitive radiotherapy. Report by the Radiation Therapy classification for lung cancer. J Thorac Oncol 2:603–612
Oncology Group. Cancer 59:1874–1881 Sakai H, Yoneda S, Kobayashi K, Komagata H, Kosaihira S,
Perez-Soler R, Fossella FV, Glisson BS, Lee JS, Murphy WK, Kazumoto T, Saito Y (2004) Phase II study of bi-weekly
Shin DM, Kemp BL, Lee JJ, Kane J, Robinson RA, docetaxel and carboplatin with concurrent thoracic radiation
Lippman SM, Kurie JM, Huber MH, Raber MN, Hong WK therapy followed by consolidation chemotherapy with
(1996) Phase II study of topotecan in patients with advanced docetaxel plus carboplatin for stage III unresectable non-
non-small-cell lung cancer previously untreated with che- small cell lung cancer. Lung Cancer 43:195–201
motherapy. J Clin Oncol 14:503–513 Saunders MI, Dische S (1990) Continuous, hyperfractionated,
Petrovich Z, Mietlowski W, Ohanian M, Cox J (1977) Clinical accelerated radiotherapy (CHART) in non-small cell carci-
report on the treatment of locally advanced lung cancer. noma of the bronchus. Int J Radiat Oncol Biol Phys 19:
Cancer 40:72–77 1211–1215
Petrovich Z, Stanley K, Cox JD, Paig C (1981) Radiotherapy in Saunders M, Dische S, Barrett A, Harvey A, Griffiths G, Palmar
the management of locally advanced lung cancer of all M (1999) Continuous hyperfractionated accelerated radio-
cell types: final report of randomized trial. Cancer 48: therapy (CHART) versus conventional radiotherapy in non-
1335–1340 small cell lung cancer: mature data from the randomised
Planting A, Helle P, Drings P, Dalesio O, Kirkpatrick A, McVie multicentre trial. Radiother Oncol 52:137–148
G, Giaccone G (1996) A randomized study of high-dose Sause WT, Scott C, Taylor S, Johnson D, Livingston R,
split course radiotherapy preceded by high-dose chemo- Komaki R, Emami B, Curran WJ, Byhardt RW, Turrisi AT,
therapy versus high-dose radiotherapy only in locally Dar AR, Cox JD (1995) Radiation Therapy Oncology Group
advanced non-small-cell lung cancer. An EORTC Lung 88-08 and Eastern Cooperative Oncology Group 4588:
Cancer Cooperative Group trial. Ann Oncol 7:139–144 preliminary results of a phase III trial in regionally
Postmus PE, Brambilla E, Chansky K, Crowley J, Goldstraw P advanced, unresectable nonsmall cell lung cancer. J Natl
(2007) The IASLC lung cancer staging project: proposals Cancer Inst 87:198–205
for the revision of the M descriptors in the forthcoming Sause W, Kolesar P, Taylor S IV, Johnson D, Livingston R,
(seventh) edition of the TNM classification for lung cancer. Komaki R, Emami B, Curran W Jr, Byhardt R, Dar AR,
J Thorac Oncol 2:686–693 Turrisi A 3rd (2000) Final results of phase III trial in
Pritchard RS, Anthony SP (1996) Chemotherapy plus radio- regionally advanced unresectable non-small cell lung can-
therapy compared with radiotherapy alone in the treatment cer: Radiation Therapy Oncology Group, Eastern Cooper-
of locally advanced, unresectable, non-small-cell lung ative Oncology Group, and Southwest Oncology Group.
cancer: a meta-analysis. Ann Intern Med 125:723–729 Chest 117:358–364
Rami-Porta R, Ball D, Crowley J, Giroux JJ, Jett J (2007) The Schaake-Koning C, van den Bogaert W, Dalesio O, Festen J,
IASLC lung cancer staging project: proposals for the Hoogenhout J, van Houtte P, Kirkpatrick A, Koolen M,
revision of the T descriptors in the forthcoming (seventh) Maat B, Nijs A (1992) Effects of concomitant cisplatin and
edition of the TNM classification for lung cancer. J Thorac radiotherapy on inoperable non-small cell lung cancer.
Oncol 2:593–602 N Engl J Med 326:524–530
Reinfuss M, Glinski B, Kowalska T, Kulpa J, Zawila K, Schild SE, Stella PJ, Geyer SM, Bonner JA, Marks RS,
Reinfuss K, Dymek P, Herman K, Skolyszewski J (1999) McGinnis WL, Goetz SP, Kuross SA, Mailliard JA, Kugler
Radiotherapy for stage III, inoperable, asymptomatic non- MD, Schaeffer PL, Jett JR (2002) Phase III trial comparing
small cell lung cancer. Final results of a prospective chemotherapy plus once-daily or twice-daily radiotherapy
randomized study (240 patients). Cancer Radiother 3: in stage III non-small-cell lung cancer. Int J Radiat Oncol
475–479 Biol Phys 54:370–378
Reynolds RD, O’Dell S (1978) Combination modality therapy Schiller JH, Harrington D, Belani CP, Langer C, Sandler A,
in lung cancer: a survival study showing beneficial results of Krook J, Zhu J, Johnson DH (2002) Comparison of four
AMCOF (Adriamycin, Metotrexate, Cyclophosphamide, chemotherapy regimens for advanced NSCLC. N Engl J
Oncobin and 5-Fluorouracil). Cancer 30:315–324 Med 346:92–98
Robertson JM, Ten Haken RK, Hazuka MB (1997) Dose Segawa Y, Kiura K, Takigawa N, Kamei H, Harita S, Hiraki S,
escalation for non small cell lung cancer using conformal Watanabe Y, Sugimoto K, Shibayama T, Yonei T, Ueoka H,
Radiochemotherapy in Locally Advanced Nonsmall-Cell Lung Cancer 431

Takemoto M, Kanazawa S, Takata I, Nogami N, Hotta K, Uematsu M, Shioda A, Tahara K, Fukui T, Yamamoto F,
Hiraki A, Tabata M, Matsuo K, Tanimoto M (2010) Phase Tsumatori G, Ozeki Y, Aoki T, Watanabe M, Kusano S
III trial comparing docetaxel and cisplatin combination (1998) Focal, high-dose, and fractionated modified stereo-
chemotherapy with mitomycin, vindesine, and cisplatin tactic radiation therapy for lung carcinoma patients. A
combination chemotherapy with concurrent thoracic radio- preliminary experience. Cancer 82:1062–1070
therapy in locally advanced non-small-cell lung cancer: Ulutin HC, Pak Y (2003) Preliminary results of radiotherapy
OLCSG 0007. J Clin Oncol 28:3299–3306 with or without weekly paclitaxel in locally advanced non-
Sekine I, Nokihara H, Sumi M, Saijo N, Nishiwaki Y, Ishikura S, small cell lung cancer. J Cancer Res Clin Oncol 129:52–56
Mori K, Tsukiyama I, Tamura T (2006) Docetaxel consol- Ulutin HC, Guden M, Oysul K, Surenkok S, Pak Y (2000)
idation therapy following cisplatin, vinorelbine, and concur- Split-course radiotherapy with or without concurrent or
rent thoracic radiotherapy in patients with unresectable stage sequential chemotherapy in non-small cell lung cancer.
III non-small cell lung cancer. J Thorac Oncol 1:810–815 Radiat Med 18:93–96
Shepherd FA, Johnston MR, Payne D, Burkes R, Deslauriers J, van Meerbeeck JP, Kramer GW, Van Schil PE, Legrand C,
Cormier Y, de Bedoya LD, Ottaway J, James K, Zee B Smit EF, Schramel F, Tjan-Heijnen VC, Biesma B,
(1998) Randomized study of chemotherapy and surgery Debruyne C, van Zandwijk N, Splinter TA, Giaccone G,
versus radiotherapy for stage IIIA non-small-cell lung European Organisation for Research and Treatment of
cancer: a National Cancer Institute of Canada Clinical Cancer—Lung Cancer Group (2007) Randomized con-
Trials Group study. Br J Cancer 78:683–685 trolled trial of resection versus radiotherapy after induction
Socinski MA, Blackstock AW, Bogart JA, Wang X, Munley M, chemotherapy in stage IIIA-N2 non-small-cell lung cancer.
Rosenman J, Gu L, Masters GA, Ungaro P, Sleeper A, J Natl Cancer Inst 99:442–450
Green M, Miller AA, Vokes EE (2008) Randomized phase Vokes EE, Gregor A, Turrisi AT (1998) Gemcitabine and
II trial of induction chemotherapy followed by concurrent radiation therapy for non-small cell lung cancer. Semin
chemotherapy and dose escalated thoracic conformal radio- Oncol 25(Suppl 4):66–69
therapy (74 Gy) in stage III non-small-cell lung cancer: Vokes EE, Herndon JE II, Crawford J, Leopold KA, Perry MC,
CALGB 30105. J Clin Oncol 26:2457–2463 Miller AA, Green MR (2002) Randomized phase II study of
Soresi E, Borghini U, Zucali R, Leoni M, Botturi M, Vergari C, cisplatin with gemcitabine or paclitaxel or vinorelbine as
Luporini G, Scoccia S (1988) A randomized clinical trial induction chemotherapy followed by concomitant chemo-
comparing radiation therapy versus radiation therapy plus radiotherapy for stage IIIB non-small-cell lung cancer:
cis-Dichlorodiamine Pltinum (II) in the treatment of locally Cancer and Leukemia Group B Study 9431. J Clin Oncol
advanced non small cell lung cancer. Semin Oncol 15(Suppl 20:4191–4198
7):20–25 White JE, Chen T, Reed R, Mira J, Stuckey WJ, Weatherall T,
Steel GG, Peckham MJ (1979) Exploitable mechanisms in O’Bryan R, Samson MK, Seydel HG (1982) Limited
combined radiotherapy–chemotherapy. Int J Radiat Oncol squamous cell carcinoma of the lung: A Southwest Oncol-
Biol Phys 5:85–91 ogy Group randomised study of radiation with or without
Taylor NA, Liao ZX, Cox JD, Stevens C, Roth J, Walsh G, doxorubicin chemotherapy and with or without levamisole
Chang JY, Guerrero T, Jeter M, Putnam J Jr, Fossella FV, immunotherapy. Cancer Treat Rep 66:1113–1120
Allen P, Komaki R (2004) Equivalent outcome of patients Whyte RI, Crownover R, Murphy MJ, Martin DP, Rice TW,
with clinical stage IIIA non-small-cell lung cancer treated DeCamp MM Jr, Rodebaugh R, Weinhous MS, Le QT
with concurrent chemoradiation compared with induction (2003) Stereotactic radiosurgery for lung tumors: prelimin-
chemotherapy followed by surgical resection. Int J Radiat ary report of a phase I trial. Ann Thorac Surg 75:1097–1101
Oncol Biol Phys 58:204–212 Yamamoto N, Nakagawa K, Nishimura Y, Tsujino K, Satouchi
Timmerman R, Papiez L, McGarry R, Likes L, DesRosiers C, M, Kudo S, Hida T, Kawahara M, Takeda K, Katakami N,
Frost S, Williams M (2003) Extracranial stereotactic Sawa T, Yokota S, Seto T, Imamura F, Saka H, Iwamoto Y,
radioablation. Results of a phase I study in medically Semba H, Chiba Y, Uejima H, Fukuoka M (2010) Phase III
inoperable stage I non-small cell lung cancer. Chest 124: study comparing second- and third-generation regimens
1946–1955 with concurrent thoracic radiotherapy in patients with
Trovo MG, Minatel E, Franchin G, Boccieri MG, Nascimben unresectable stage III non-small-cell lung cancer: West
O, Bolzicco G, Pizzi G, Torretta A, Veronesi A, Gobitti C Japan Thoracic Oncology Group WJTOG0105. J Clin Oncol
(1992) Radiotherapy versus radiotherapy enhanced by 28:3739–3745
cisplatin in stage III non small cell lung cancer. Int J Yorke E (2001) Advantages of IMRT for dose escalation in
Radiat Oncol Biol Phys 24:11–15 radiation therapy for lung cancer. Med Phys 28:1291–1294
Tri-Modality Therapy in Locally Advanced
Non-Small-Cell Lung Cancer
Jan P. van Meerbeeck and Elke Vandenbroucke

Contents Abstract
This review addresses recent evidence on the role
1 Introduction.............................................................. 434 of surgery in different clinical subsets of so called
2 Stage III Without Mediastinal Lymph Node ’potentially resectable’ stage III NSCLC. In some
Involvement .............................................................. 434 selected subsets of stage III patients, in particular
3 Stage III with Mediastinal Lymph Node those with T3-4N0-1, surgery plays a key role.
Involvement .............................................................. 437 Resection has to be decided on by a multidisci-
plinary team and carried out in a high volume
4 Discussion and Future Directions.......................... 440
institution by an experienced team. In all patients
5 Conclusion ................................................................ 442 who are being considered for resection, accurate
References.......................................................................... 442 staging of the mediastinum and excluding occult
metastasis is recommended. Comorbidities of
patients have considerable influence on the overall
treatment plan in each individual patient. The
preferred mode of treatment for locally advanced
NSCLC with clinical mediastinal lymph node
invasion is a combined modality treatment with
concurrent chemo- and radiotherapy.

Keywords
Chemoradiotherapy  Combined modality  Non-
small cell lung cancer  Stage III  Surgery

Abbreviations
J. P. van Meerbeeck (&)
Department of Respiratory Medicine, NSCLC Non-small cell lung cancer
University Hospital Ghent, OS Overall survival
De Pintelaan 185, 9000 Ghent, Belgium PFS Progression-free survival
e-mail: jan.vanmeerbeeck@ugent.be SST Superior sulcus tumor
J. P. van Meerbeeck R0 Complete resection
Department of Respiratory Medicine, pCR Pathological complete response
Lung Oncological Network Ghent University Hospital,
Ghent, Belgium
E. Vandenbroucke
Department of Respiratory Medicine,
Monica Hospital, Antwerp, Belgium

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 433
DOI: 10.1007/174_2011_306, Ó Springer-Verlag Berlin Heidelberg 2011
434 J. P. van Meerbeeck and E. Vandenbroucke

Table 1 Definition of clinical subsets in stage III NSCLC according to the current and the proposed new TNM-classification
Description TNM (Mountain 1997) Stage Proposed TNM Stage
(Goldstraw et al. 2007)
Without mediastinal lymph node involvement
Chest wall invasion including SST or tumor T3N1 IIIA T3N1 IIIA
\2 cm distal to main carina
Central tumor with local invasion T4N0-1 IIIB T4N0-1 IIIA
Same lobe nodules T4N0-1 IIIB T3N0 IIB
T3N1 IIIA
Malignant pleural effusion T4N0-1 IIIB M1a IV
With mediastinal lymph node involvement
Ipsilateral mediastinal nodes T1-3N2 IIIA T1-3N2 IIIA
Same lobe nodules T4N2 IIIB T3N2 IIIA
Central tumor with local invasion T4N2 IIIB
Contralateral or supraclavicular nodes T1-4N3 IIIB T1-4N3 IIIB
TNM tumor-node-metastasis, SST superior sulcus tumor

1 Introduction 2 Stage III Without Mediastinal


Lymph Node Involvement
Lung cancer has become the leading cause of death
among cancers worldwide. Non-small-cell lung Superior sulcus tumors (SST) are a distinct group
cancer (NSCLC) accounts for approximately 85% of among NSCLC because of their localization and
all cases and has a poor overall 5-year survival of clinical presentation. A combined modality treatment
about 15% (Molina et al. 2008). At the time of pre- was first presented with the results of preoperative
sentation, at least 40% of patients are diagnosed in an radiotherapy followed by resection (Shaw et al.
advanced stage and about one-third have locally- 1961). During the following decades, this approach
advanced (stage III) disease, which is a heterogeneous remained the standard of care with 5-year survival
group of patients ranging from either T3N1 over rates up to 30%, but less than 10% for patients with
advanced T4 tumors, positive mediastinal lymph N2-disease. Loco-regional recurrence was the com-
nodes or malignant pleural effusion in the current monest cause of relapse, emphasizing the need for
TNM-classification (Mountain 1997; Goldstraw et al. better loco-regional treatment strategies.
2007). The standard of care for most patients is a Two multi-centric phase 2 trials (Rusch et al. 2007;
combined modality treatment, wherein the role of Kunitoh et al. 2008) have recently shown high
surgery remains controversial. resectability, local control rates and survival using a
This review addresses recent published evidence tri-modality regimen consisting of induction plati-
on the role of surgery in different clinical subsets of num-based chemoradiotherapy followed by surgery
so-called ‘potentially resectable’ stage III NSCLC (Table 2).
(Table 1). A discussion of the different induction The Southwest Oncology Group (SWOG) (Rusch
regimens is beyond the limits of this review. et al. 2007) treated 110 patients with T3-4 SST without
The evidence was retrieved from ‘PubMed’ using mediastinal lymph node involvement with two cycles of
the keywords (surgery), (non-small-cell lung cancer) cisplatin-etoposide and concurrent radiation. After
and limited to original reports published from 2007 induction, of 95 patients without progression, 88 under-
onward. The articles were then selected manually for went thoracotomy. All patients, whether or not resected,
further appropriateness. received two additional cycles of chemotherapy. Median
Table 2 Recent evidence for surgery in superior sulcus, central T4 and multifocal T4 NSCLC
Author Setting Stage Analyzed Regimen Pneumo- Exploratory Clinical R0 Pathological pCR Treatment Median 2-3-5-year PFS
populations nectomy thoracotomy response resection downstaging (%) mortality survival survival (%) (mo)
(n) (n) (%) (%) (%) (%) (mo)
Superior sulcus tumors
Rusch et al. Phase 2 trial cT3-4 ITT 2 cycles CE ? RT — — 42 76 — 29 4.5 33 NR-NR- 44 —
(2007) N0-1 (n = 110) (45 Gy) ? surgery
SWOG ? 2 cycles CE
PP 3 2 43 — 33 36 — 94 (R0) NR-NR- 55 —
9416
(n = 88) (R0)
Intergroup
trial 0160
Kunitoh Phase 2 trial cT3-4 ITT 2 cycles MVP ? RT — — 61 68 — 16 4 — NR-62-56 28
et al. (2008) N0-1 (n = 75) (45 Gy) ? surgery
JCOG ± boost RT (21.6 Gy) if
PP 0 1 — — 40 — — — NR-NR- 70 —
9806 unresectable disease
(n = 57) (R0)
Marra et al. Prospective cT3-4 ITT 3 cycles CE or — — 48 — — — 6 54 74-NR- 46 52
(2007) serie N0-3 (n = 31) CP ? 1 cycle (5-
CE ? 45 Gy year)
? surgery
PP 1 0 — 100 38 45 — — — —
(n = 29)
Pourel et al. Prospective cT3-4 ITT 2 cycles CE + 45 Gy ? — — NR — — — — 26.7 55-NR-40 15.2
(2008) serie N0-2 (n = 107) surgery
± adjuvant treatment
PP 21 1 — 90 60 39 6 36.4 62-NR-51 20.9
(n = 72)
Yildizeli Retrospective cT4 ITT Neoadjuvant — — NR — NR — 1 28 NR-NR- 37 —
et al. (2008) serie N0-1 (n = 126) treatment ? surgery
± adjuvant treatment
PP 3 0 — 90 — NR — — NR-NR- 39 —
(n = 126) (R0)
Tri-Modality Therapy in Locally Advanced Non-Small-Cell Lung Cancer

Author Setting Extension No. cN0-1 (%) cN2-3 Pneumo- Neoadjuvant Adjuvant R0 pCR 30 day Median 5-year survival according to nodal status
patients (%) nectomy (n) treatment (%) treatment resection (%) mortality survival
pN0-3 (%) pN0 (%) pN1 (%) pN2-3 (%)
(%) (%) (%) (mo)
Central T4 tumors
Yildizeli Retrospective Carina 92 87 23 29 98 — 6 28 42 40 47 18
et al. (2008) serie SVC 39 100 0 32 36 59 85 — 8 19 29
Mediastinum 14 12 43 64 86 — 7 37 61
De Leyn Prospective PA 5
et al. (2009) serie LA 5 100 0
Aorta 3 ITT n = 32 — 100 100 78 NR — NR 74 — — —
Mediastinum 3 PP n = 27 11 100 100 93 7 0 NR 77 (R0) — — —
Subclavian artery 1
SST 15
Misthos Retrospective Aorta 13 100 0 5 0 10 100 — 0 38 31 100 37 0
et al. (2007) serie SVC 9 100 0 0 0 100 NR — 0 31 11 NR NR NR
Farjah et al. Retrospective Not specified 1177 68 22 20 4–8 40–55 NR NR 10 NR 20 28 NR 8
(2008) serie
Wu et al. Retrospective PV or LA 46 93 7 30 22 100 NR NR 0 NR 22 29 28 18 (3-year)
(2009) serie
Anraku Retrospective Spine 23 NR NR 1 100 NR 83 43 9 47 58 (3-year) NR NR NR
et al. (2009) serie 72 (R0)
435

(continued)
436 J. P. van Meerbeeck and E. Vandenbroucke

PFS progression free survival, R0 microscopically radical resection, pCR pathological complete response, CE cisplatin-etoposide, CP cisplatin-paclitaxel, RT radiotherapy, NR not reported, MVP mitomycin ? vindesine or vinblastine ? cisplatin,
survival for all eligible patients was 33 months versus

pN+
5-year survival

(%)

NR
94 months for the patients who underwent complete


30
47
according to
nodal status
pN0
resection (R0). Pathological complete response (pCR)
(%) was a significant prognostic factor for survival. Relapse

48

58

64
was predominantly distant with the brain as the
commonest site.
survival (%)

The Japanese Clinical Oncology Group (JCOG)


5-year



48

57
(Kunitoh et al. 2008) included 75 patients with T3-4
N0-1 tumors from 19 centers. Of the patients who
survival (mo)

started induction treatment, 57 (76%) underwent sur-


gical resection. Fiftyone (68%) patients had a R0
Median

PP per protocol (operated patients), ITT intention-to-treat population, PV pulmonary vein, LA left atrium, SVC superior vena cava, PA pulmonary artery, SST superior sulcus tumor, mo months
45.4
22.8

resection with pCR in 16%. In un-resected or incom-


44

68

pletely resected cases, boost radiotherapy was admin-


mortality (%)

istered. The 3- and 5-year overall and progression-free


survival rates were 61, 56, 49, and 45%, respectively.
30 day

NR

In contrast to the SWOG trial (Rusch et al. 2007),


4
0

subset analysis revealed that patients with clinical


pN2/3

T3-disease had a better outcome than those with clin-


(%)

100
49
0
7

ical stage T4. The clinical N-stage and histologic type


of the tumor did not significantly affect the overall
[1 satellite

survival (OS) or progression-free survival (PFS). As


lesion (%)

expected, the survival rate was good in patients in


NR
30
19

whom R0 resection could be achieved with a projected


5-year OS of 70% compared to 24% in incompletely
resected patients. For un-resected or incompletely
lesion (%)
1 satellite

resected cases, loco-regional relapse was predominant.


NR
32
81

91

For completely resected cases, relapse at distant sites


was the most frequent pattern of recurrence with some
R0 resection

patients only relapsing in the brain.


(%)

100
100

NR

The results of both trials have further been confirmed


86

by other recent prospective and retrospective series


(Table 2) (Marra et al. 2007; Pourel et al. 2008; Yildizeli
nectomy (n)
Pneumo-

et al. 2008; De Leyn et al. 2009). A tri-modality


NR
12

approach is feasible in most series with a reasonably low


0

mortality and tolerable morbidity rate. The survival data,


cT4
(%)

NR

with a 5-year OS rate in the intention-to-treat population


15

51
0

of 44% in the US trial and 56% in the Japanese trial, are


patients

clearly superior to the historical value of 30%. Most if


No.

56
67
53

35

not all series show the following factors to be predictive


of success: R0 resection, lobectomy, pCR and absence of
pT4N+
pT4N0
Stage

cT4/

cT4/
pT4

pT4

invasion in either the intervertebral foramina, subclavian


artery or brachial plexus. Ipsilateral supraclavicular
Retrospective
Retrospective

Retrospective

lymph node involvement, only specified in the JCOG,


Table 2 (continued)
Setting

was not prognostic. Other variables such as age, gender,


serie
serie

serie

performance status or histological subtype had no


Multifocal T4 tumors

influence on survival.
Trousse et al.

Although there are no formal randomized trials,


Port et al.

Rao et al.
Author

preoperative chemoradiotherapy followed by surgical


(2008)
(2007)

(2007)

resection is now regarded as the optimal management


Tri-Modality Therapy in Locally Advanced Non-Small-Cell Lung Cancer 437

for locally-invasive tumors of the superior sulcus in specimen (n = 45). Patients with multi-centric bron-
carefully selected patients without mediastinal lymph cho-alveolar cancer (BAC) and those with different
node involvement. histologies in the primary versus the satellite lesions
Patients with central T4 tumors have invasion of were excluded. Five-year OS was 48% for all patients
either the heart, great vessels, trachea, esophagus or and 58% in patients without nodal involvement.
vertebrae (Mountain 1997). Most patients have also The number of satellite lesions in the resected lobe did
mediastinal lymph node involvement. They are often not appear as a prognostic factor.
considered unresectable and treated with chemoradia- Trousse et al. (2008) analyzed 56 patients with
tion, as is generally recommended for patients with postoperatively detected multi-focal NSCLC. Overall
stage IIIB NSCLC (Shen et al. 2007; Jett et al. 2007). 5- and 10-year survival rates were 48 and 30%
Although there are no randomized data, cumulative respectively. The tumors with nodal involvement
evidence suggests that selected T4 tumors without (N1-3) had a 5- and 10-year survival of 30 and 18%.
mediastinal lymph nodes can be resected with better The better results in the series from Rao et al. (2007)
long-term outcome than historically-associated with may be related to the high percentage of BAC in this
stage IIIB NSCLC as shown in Table 2 (Yildizeli et al. study. Adenocarcinoma or BAC, R0 resection, node-
2008; De Leyn et al. 2009; Misthos et al. 2007; Farjah negative patients and the absence of vascular invasion
et al. 2008; Wu et al. 2009; Anraku et al. 2009). The are frequently reported prognostic factors for better
largest retrospective cohort study (Anraku et al. 2009) survival in these surgical series of multi-focal T4.
reports on 1,177 operated patients with T4 tumors with Patients with intralobar multi-focal NSCLC hence
a 20% 5-year survival rate, although they were not seem to have a more favorable prognosis after sur-
stratified according to their localization. The largest gical resection than might be predicted by their T4
experience of resection for central T4 tumors involves stage. 5-year survival rates, especially in N0 patients,
carinal resection usually together with right pneumo- approximate those with stages IB of II NSCLC. These
nectomy. Yildizeli et al. (2008) performed 109 carinal results support the proposal of the forthcoming new
resections in patients with carinal (n = 92) or superior TNM-classification wherein ipsilobar multi-focal T4
vena cava (n = 17) involvement. Median OS time was disease will be recoded as T3 (Goldstraw et al. 2007).
28 months and 5-year survival 42.5%. In patients with
N0/N1 disease 5-year OS was 50%. Wu et al. (2009)
reported 46 patients with tumors invading the base of 3 Stage III with Mediastinal Lymph
the pulmonary vein or the left atrium (LA). Partial LA Node Involvement
resection was performed in all patients with an overall
5-year survival rate of 22% and a worse prognosis with Patients with positive mediastinal lymph nodes are
N2-involvement. A retrospective analysis of 23 patients the largest group within stage III NSCLC. The extent
undergoing radical vertebrectomy after induction che- of mediastinal node involvement has an inverse cor-
moradiation for NSCLC invading the spine (including relation with survival. Although the current staging
SST) reports a median OS of 47 months and 3-year system does not divide the N2-category into subsets
survival of 58% (Anraku et al. 2009). Patients who based on the size of mediastinal disease, this factor
achieved (near) pCR demonstrated significantly better must be considered when evaluating the results of
survival than those who did not (3-year survival 92 clinical series. The subgroup with unforeseen
versus 20%). N2-involvement, incidentally found on final patho-
NSCLC with multiple intralobar lesions represents logic examination (IIIA-1) or recognized intra-oper-
either a multi-focal origin or intralobar metastasis. atively (IIIA-2), accounts for 14 - 24% of patients.
Several retrospective series have shown that , patients However, the largest subgroup of stage IIIA (67%)
in whom multi-focal T4 was found at resection consists of patients with clinical single or multiple
behave differently than T4 cases with other extension. level ipsilateral lymph node invasion (IIIA-3), or
The following series confirm these findings (Table 2). ‘bulky’ N2-disease at imaging (IIIA-4). Preopera-
Port et al. (2007) reviewed 53 patients with resected tively proven stage IIIA-3 disease is variably con-
lung cancer containing intralobar satellite lesions sidered ‘resectable’ with or without induction or
detected preoperatively (n = 8) or in the resected adjuvant therapy, whereas patients with stage IIIA-4
Table 3 Outcome of surgery as part of combined modality treatment in stage III-N2/3 NSCLC
438

Author Setting Stage Analyzed Regimen Pneumo- Exploratory Clinical R0 Nodal pCR Mortality Median 2-3-5- Median
populations nectomy thoracotomy response resection downstaging (%) rate (%) OS year PFS
(%) (%) (%) (%) (%) (months) survival (months)
(%)
PP/ITT PP PP ITT PP PP PP PP ITT ITT ITT

Garrido Phase 2 IIIA(N2) 46/69 Three cycles cisplatin- 39 11 56 72 27(R0) 3(R0) 8 15.6– NR-37- 9.9
et al. 2007 gemcitabine- 36.9 (R0) 21
(2007) docetaxel ? surgery
IIIB 44/67 43 23 56 66 – 24(R0) 7 16.8–
SLCG 9901
(T4N0-1) 60.6(R0)
Van Phase 3 III-N2 154/167 I: three cycles platinum- 47 14 612 50 41 5 9 16.4 35-NR- 9.0
Meerbeeck based combined 16
et al. CT ? surgery
(2007)
154/165 II: three cycles platinum- – – 612 – 0 – \1 17.5 41-NR- 11.3
EORTC
based combined CT ? RT 14
08941
(60 Gy)
Thomas Phase 3 IIIA/B 142/264 I: three cycles CE ? bid RT 35 8 50 37 46 60 9 15.7 NR-28- 9.5
et al. (45 Gy) ? carbo- 21
(2008) vindesine ? surgery
GLCCG
154/260 II: three cycles 35 8 46 32 29 20 5 17.6 NR-26- 10
CE ? surgery ? 18
conventional RT (54 Gy)
Gottfried Phase 3 IIB(T3N0) 107/155 I: three cycles 39 8 53 74 29 5 3 32.3 59-47- 16.8
et al. NIP ? surgery (n = 42) NR
(2008)1
IIIA- II: three cycles 31.8 62-49- 16.8
B(T4N0) NIP ? surgery ? 2 cycles NR
NIP (n = 37)
Uy et al. Retrospective IIIA(N2) 40 Two cycles CE ? RT 27 0 NR 92 NR 17 7 40 NR-52- 37.1
(2007) serie (45 Gy) ? surgery ? NR
2 cycles CE
Mansour Retrospective IIIA-N2 28 I: induction CT ? persistent 11 28 NR–NR- NS
et al. serie N2 ? surgery 32
(2008)
32 II: induction CT ? 100 0 NR NR NR NR 12 27 NR–NR- NS
pN0-1 ? surgery 35
93 III: surgery alone 10 15 NR–NR- NS
12
Carretta Retrospective IIIA/B 110 MVP ? surgery ? ± RT 27 0 84 100 NR 12 2 NR NR–NR- NR
et al. serie 50–56 Gy 46
(2008)
GC ? surgery ?
± chemotherapy
Yap et al. Retrospective IIIA/B 25/33 Three cycles PG ? RT 18 0 70 100 NR 24 \1 29.9 NR–NR- 18.3
(2008) serie (50 Gy) ? surgery 36
1
study was closed prematurely because of low inclusion rate;
2
n = 579
PFS progression-free survival, OS overall survival, R0 microscopically radical resection, pCR pathologic complete response, CT chemotherapy, CE cisplatin-etoposide, RT radiotherapy, NR not reported, NS not significant, MVP mitomycin ? vindesine
or vinblastine ? cisplatin, GC gemcitabin–cisplatine, NIP vinorelbine–ifosfamide–cisplatin, PG paclitaxel–carboplatin, PP per protocol (operated patients), ITT intention-to-treat population (registered/randomozed patients)
J. P. van Meerbeeck and E. Vandenbroucke
Tri-Modality Therapy in Locally Advanced Non-Small-Cell Lung Cancer 439

and IIIB-N3 are considered ‘primary unresectable’ radiotherapy arm and predominantly distant in the
(Jett et al. 2007; Robinson et al. 2007). surgery arm. Half of patients however received
The recently published evidence with regard to the postoperative radiotherapy, which may have influ-
surgical treatment in stage III NSCLC with medias- enced the local relapse rate. In a post-hoc unplanned
tinal lymph node involvement is summarized in subgroup analysis of the surgical arm, 5-year survival
Table 3. The Spanish Lung Cancer Group (SLCG) was longer if there was a nodal downstaging to N0 or
performed a phase 2 trial of induction chemotherapy if lobectomy was performed (Van Schil et al. 2005).
with a cisplatin-based triplet followed by surgery for The authors concluded that, after a radiologic
stage III-N2 and selected IIIB (T4N0-1) (Garrido et al. response to induction chemotherapy, surgery was not
2007). 136 patients were included of whom 129 were superior to radiotherapy. In the surgery arm, the
assessable for treatment. Only 90 patients (70%) extent (lobectomy versus pneumonectomy) and
underwent thoracotomy and R0 resection was achieved the type (R0 versus R1) of resection were prognostic.
in 62 patients. Postoperative treatment was allowed in In the radiotherapy arm, none of the patient charac-
patients with pathologically persistent N2-disease, teristics was prognostic. In view of its lower mortality
unresectable disease or incomplete resection. Median and morbidity, sequential chemoradiotherapy was
OS for both stages III-A and III–B by intention-to-treat recommended to remain the standard of care in
analysis was 15.9 months and the median PFS was patients with initially ‘unresectable’ IIIA-N2 disease.
9.9 months. There was no significant difference in The randomized phase 3 trial by the German Lung
median OS between stage IIIA and IIIB. In this study Cancer Cooperative Group (Thomas et al. 2008) com-
clinical response, R0 resection and nodal down-staging pared the intensity of the induction regimen: neoadju-
in stage IIIA-N2 were significant prognostic factors for vant concurrent chemoradiation versus chemotherapy
survival. The type of surgical procedure (e.g. pneu- alone. In the latter surgery was followed by radiother-
monectomy) was not correlated with outcome. apy up to 54 Gy. The severe toxicity and mortality rates
Two large multi-center randomized phase 3 trials were significantly higher in the group of patients with
have explored the role of surgery versus radiotherapy preoperative radiotherapy with almost a doubling in the
after induction treatment for stage III-N2 NSCLC. postoperative mortality rate (9 versus 4.5%) especially
The first study was already reported by the North after pneumonectomy. Despite the finding that the
American Intergroup in 2005, but is not published yet more intense induction treatment resulted in a higher
(Albain et al. 2009). pCR rate, this did not translate in a better outcome,
In the EORTC 08941 trial (Van Meerbeeck et al. casting a doubt on the value of pCR as a surrogate
2007), 582 patients with stage IIIA-3 and IIIA-4 were marker of outcome. In this study, 67% of patients had
included. ‘Un-resectability’ was defined as any stage IIIB. The criteria for ‘unresectable disease’
mediastinal lymph node invasion by a non-squamous before and after induction treatment seemed not well
NSCLC or in case of squamous cell cancer, defined, and the comparison of the two induction reg-
involvement of more than one station. In case of imens—chemotherapy versus chemoradiotherapy—
response on chest CT-scan after induction chemo- may be blurred by several other differences between the
therapy, patients were randomized between surgery two groups: standard fractionation versus hyper-frac-
and radiotherapy. Postoperative radiotherapy was tionation and preoperative versus postoperative radio-
allowed in the surgical arm when resection was not therapy. Additionally, the use of hyper-fractionated
radical. The primary endpoint was OS analyzed radiotherapy in NSCLC has not been shown to improve
according to the intention-to-treat principle. Overall outcome (Belani et al. 2005).
response rate to induction chemotherapy was 61%, The phase 3 trial by Gottfried et al. (2008),
however this response was not pathologically con- assessing the role of adjuvant chemotherapy in
firmed. The R0 resection rate was 50 with 5% pCR. patients with stage IIB-IIIA/B, was closed prema-
Downstaging to N0-1 was obtained in 41%. Median turely because of a low inclusion rate. Of 107 patients
OS (17.5 versus 16.4 months) and 5-year survival operated, 37 were randomized to the adjuvant treat-
rates (14 versus 15.7%) as well as PFS for patients in ment arm and 42 to the observational arm. There were
the surgery versus radiotherapy arm were similar. Site no differences in outcome, although the data have to
of first relapse was more often loco-regional in the be interpreted with caution as the study was not
440 J. P. van Meerbeeck and E. Vandenbroucke

stratified by stage. Response rates and pCR were metastasis, sometimes only the brain, is the com-
comparable to the above-mentioned results. monest form of relapse in completely resected
Impressive results of induction chemoradiation patients, improved systemic therapy will be needed.
before surgical intervention for selected patients with There is no real consensus about the length of induction
stage IIIA-N2 NSCLC were recently published by Uy chemotherapy nor about the need of any adjuvant
et al. (2007), who adopted the protocol from the treatment or not. Whereas in the SWOG trial every
surgical arm of the North American Intergroup trial. patient received additional chemotherapy, postopera-
Other retrospective surgical series confirm 5-year tive chemotherapy or radiotherapy was administered in
survival rates ranging from 30–45% after induction most studies in case of unresectability or incomplete
chemoradiotherapy (Table 3). The results from resection, without resulting in a better 5-year survival.
Mansour et al. (2008) suggest that pneumonectomy is Even the role of postoperative radiotherapy in incom-
justified even in patients with persistent N2-disease pletely resected patients is uncertain as the major
after induction chemotherapy. Comparing the series relapse is still locoregional. Prophylactic cranial irra-
of Carretta et al. (2008) and Yap et al. (2008), there is diation is to be considered as part of clinical trials in
no benefit from chemoradiotherapy over chemother- other subsets of locally-advanced NSCLC.
apy alone as induction treatment. As in previous There are only limited data available, but in selected
trials, patients with R0 resection, nodal down-staging patients with clinical T4 N0-1 NSCLC with either
and pCR seemed to benefit most from surgery, multi-focal ipsilobar disease or with central extension
although there are no randomized data available in to the main carina, great vessels or vertebral column,
these subgroups. upfront surgery should be considered. There is cur-
The role of surgery after induction chemotherapy rently no role for induction therapy. The SLCG
in patients with stage IIIB-N3 NSCLC remains (Garrido et al. 2007) included patients with T4N0-1
unclear. No phase 3 data has presently shown that a NSCLC in a phase 2 trial but showed no survival benefit
neoadjuvant treatment followed by surgery results in in this subset of patients after post-induction surgery
prolonged survival compared to adequate chemora- compared to other series with central or multi-focal T4
diation only. tumors not receiving induction therapy (Misthos et al.
2007; Farjah et al. 2008; Wu et al. 2009). In the series of
Farjah et al. (2008), there was no apparent relation
4 Discussion and Future Directions between neoadjuvant therapy and short- or long-term
outcome. This finding is in contrast with the results
According to recent guidelines (Shen et al. 2007; Jett reported in SST. One explanation is that neoadjuvant
et al. 2007; Robinson et al. 2007; D’Addario and therapy may not improve the ability to achieve R0
Felip 2008; NCCN Clinical Practice Guidelines in resection in patients with T4 disease. Of interest is the
Oncology. Non-small cell lung cancer.V.2 2009), lack of data reporting the impact of any induction
there are now accepted indications for surgery in treatment in lowering the pneumonectomy rate in these
selected subsets of patients with stage III NSCLC, series, which might well decrease their operative
especially as part of a combined modality treatment. mortality rate. Postoperative treatment should be
Selected patients with SST with good performance dictated by the pathological findings and include post-
and without mediastinal lymph node involvement are operative radiotherapy in case of incomplete resection or
preferentially treated with combined chemoradiation chemotherapy in case of unforeseen hilar or mediastinal
induction therapy followed by surgery. Recent lymph node invasion. Due to their low prevalence, it is
promising results confirm that tri-modality is feasible highly unlikely that a randomized trial will ever be
with an acceptable low morbidity and mortality rate. conducted comparing the role of surgery to a non-sur-
The resectablility depends on the localization and the gical approach only in these subsets of stage III NSCLC
anatomical extension of the tumor—either in the without mediastinal lymph node invasion. As there are
anterior, middle or posterior compartments—which no randomized controlled data available, the reported
require a different surgical approach (Girard and role of surgery is mainly based on the centers’ expertise.
Mornex 2007; Shaw et al. 1961; Dartevelle et al. It is hence crucial that these patients are evaluated and
1993; Grunenwald and Spaggiari 1997). As distant treated by expert multi-disciplinary teams.
Table 4 Ongoing phase 3 clinical trials including surgery in stage III NSCLC
Title of study Design Schedule Primary Estimated Start Study Collaborators Country
objective No. status
Patients
Surgery versus no surgery
Neoadjuvant Phase I: chemotherapy ? radiotherapy Overall 406 January Recruiting Rigshospitalet Denmark
chemotherapy ± surgery in 3 survival 1998 Denmark
NSCLC stage IIIA-N2 II: chemotherapy ? surgery ?
(Scandinavian Neoadjuvant Phase radiotherapy
III Study of Induction 2009)
Standard definitive boost Phase I: chemotherapy (paclitaxel- Overall 500 July Recruiting Essen Germany
radiotherapy versus surgery 3 cisplatin) ? radiochemotherapy survival 2003 Paris
following induction chemotherapy (cisplatin-vinorelbine ? 45 Gy Tübingen
and neoadjuvant bid) ?boost radiotherapy ?
radiochemotherapy based on chemotherapy
hyper-fracionated accelerated II: chemotherapy (paclitaxel-
Tri-Modality Therapy in Locally Advanced Non-Small-Cell Lung Cancer

radiotherapy in NSCLC IIIA-N2/ cisplatin) ? radiochemotherapy


III-B ESPATÜ-trial-GCS 2.1 (cisplatin-vinorelbine ? 45 Gy
bid) ? surgery
Sequential chemoradiotherapy versus chemotherapy as induction treatment
Chemotherapy ± radiation Phase I: chemotherapy (cisplatin- Event-free 120 April Recruiting SAKK Switzerland
therapy before surgery in treating 3 docetaxel) ? surgery survival 2001 NCI
patients with NSCLC stage IIIA- II: chemotherapy (cisplatin- Operability
N2 (Preoperative docetaxel) ? radiation ? surgery after
Chemoradiotherapy versus chemotherapy
Chemotherapy Alone 2009)
441
442 J. P. van Meerbeeck and E. Vandenbroucke

In stage III-N2/3 NSCLC, the role of surgery a multi-disciplinary team and carried out in a high
compared to radical and adequate modern thoracic volume institution by an experienced team.
radiotherapy for local control after induction treat-
ment is still a challenge. There is no consensus on the
intensity of induction therapy. In patients receiving References
induction chemoradiation, early toxicities (e.g.
esophagitis and hematotoxicity) are increased and in Albain KS, Swann RS, Rusch VW et al (2009) Radiotherapy
those undergoing surgery, post-operative mortality plus chemotherapy with or without surgical resection for
rate is higher especially when a pneumonectomy is stage III non-small-cell lung cancer: a phase III randomised
controlled trial. Lancet 374(9687):379–386
performed. Further randomized controlled trials are
Anraku M, Waddell TK, de Perrot M et al (2009) Induction
needed to show the superiority of neoadjuvant treat- chemoradiotherapy facilitates radical resection of T4
ment followed by surgical resection for patients with non-small cell lung cancer invading the spine. J Thorac
stage III-N2/3 NSCLC. Two large randomized con- Cardiovasc Surg 137:441–447
Belani CP, Wang W, Johnson DH et al (2005) Phase III study of
trolled trials (Table 4) are on-going in stage IIIA-N2
the Eastern Cooperative Oncology Group (ECOG 2597):
(Scandinavian Neoadjuvant Phase III Study of induction chemotherapy followed by either standard thoracic
Induction 2009) and IIIA/B NSCLC (ESPATÜ, radiotherapy of hyperfractionated accelerated radiotherapy
Eberhardt, personal communication). Whether che- for patients with unresectable stage IIIA and B non-small cell
moradiation is superior to chemotherapy alone as lung cancer. J Clin Oncol 23:3760–3767
Carretta A, Ciriaco P, Melloni G et al (2008) Results of surgical
induction therapy, is currently investigated in a treatment after neoadjuvant chemotherapy for stage III
Swiss trial (Preoperative Chemoradiotherapy vs. non-small cell lung cancer. World J Wurg 32:2636–2642
Chemotherapy Alone 2009). D’Addario G, Felip E (2008) Non-small-cell lung cancer:
R0 resection, nodal down-staging and pCR are all ESMO clinical recommendations for diagnosis, treatment
and follow-up. Annals Oncol 19((Supplement)):ii39–ii40
prognostic factors for survival in patients with stage III Dartevelle PG, Chapelier AR, Macchiarini P et al (1993)
NSCLC. However, these are post hoc variables deter- Anterior transcervical-thoracic approach for radical resec-
mined on resection specimen, and they are not helpful tion of lung tumors invading the thoracic inlet. J Thorac
to the clinician’s preoperative decision-making. Whe- Cardiovasc Surg 105:1025–1034
De Leyn P, Stroobants S, De Wever W et al (2006) Prospective
ther surgery should be reserved only for patients with comparative study of integrated positron emission tomog-
mediastinal clearance after induction chemotherapy is raphy-computed tomography compared with remediastinos-
unclear. FDG-PET/CT-scan is of great interest, how- copy in the assessment of residual mediastinal lymph node
ever the sensitivity in the detection of residual medi- disease after induction chemotherapy for mediastinoscopy-
proven stage IIIA-N2 non-small cell lung cancer: a Leuven
astinal lymph node disease is low. Post-induction Lung Cancer Group Study. J Clin Oncol 24:3333–3339
mediastinoscopy has been reported to have a disap- De Leyn P, Vansteenkiste J, Lievens Y et al (2009) Survival
pointing low sensitivity (De Leyn et al. 2006). after trimodality treatment for superior sulcus and central
T4 non-small cell lung cancer. J Thorac Oncol 4:62–68
Farjah F, Wood DE, Varghese TK et al (2008) Trends in the
operative management and outcomes of T4 lung cancer.
Ann Thorac Surg 86:368–375
5 Conclusion Garrido P, Gonzales-larriba JL, Insa A et al (2007) Long term
survival associated with complete resection after induction
chemotherapy in stage IIIA (N2) and IIIB (T4N0–1) non-
The preferred standard of care for most locally- small cell lung cancer patients: the Spanish Lung Cancer
advanced NSCLC is a combined modality treatment Group Trial 9901. J Clin Oncol 25:4736–4742
with chemotherapy and radiotherapy. However, there Girard N, Mornex F (2007) Therapeutic strategies in
are some selected subsets of stage III patients, in superior sulcus tumors: a model for combined modality
therapy in non-small cell lung cancer. Cancer/radiothéra-
particular those with T3-4N0-1, in whom surgery pie 11:59–66
plays a key role. In all patients who are being Goldstraw P, Crowley JJ, Chansky K et al (2007) On behalf of
considered for resection, accurate staging of the the International Association for the study of Lung Cancer
mediastinum and excluding occult metastasis is rec- International Staging Committee and Participating Institu-
tions. The IASLC Lung Cancer Staging Project: proposals
ommended. Comorbidities of patients have consider- for the revision of the TNM stage groupings in the
able influence on the overall treatment plan in each forthcoming (seventh) edition of the TNM classification of
individual patient. Resection has to be decided on by malignant Tumors. J Thor Oncol 2:705–714
Tri-Modality Therapy in Locally Advanced Non-Small-Cell Lung Cancer 443

Gottfried M, Ramlou R, Krzakowski M et al (2008) Cisplatin- evidence-based clinical practice guidelines (2nd edition).
based three drugs combination (NIP) as induction and Chest 132:243–265
adjuvant treatment in locally advanced non-small cell lung Rusch VW, Giroux DJ, Kraut MJ et al (2007) Induction
cancer. final results. J Thorac Oncol 3:152–157 chemoradiation and surgical resection for superior sulcus
Grunenwald D, Spaggiari L (1997) Transmanubrial osteomus- non-small cell lung carcinomas: long-term results of
cular sparing approach for apical chest tumors. Ann Thorac Southwest Oncology Group Trial 9416 (Intergroup Trial
Surg 63:563–566 0160). J Clin Oncol 25:313–318
Jett JR, Schild SE, Keith RL et al (2007) Treatment of non- Scandinavian neoadjuvant Phase III study of induction chemo-
small cell lung cancer, stage IIIB: ACCP evidence-based therapy followed by irradiation alone or surgery plus
clinical practice guidelines (2nd edition). Chest 132: irradiation in NSCLC stage IIIA/N2 (T1N2, T2N2, T3/
266S–276S N2). NCT00273494. Bethesda MD: National Library of
Kunitoh H, Kato H, Tsuboi M et al (2008) Phase II trial of Medicine, 2009. (Accessed Feb 5, 2011 at http://www.
preoperative chemoradiotherapy followed by surgical resec- clinicaltrials.gov)
tion in patients with superior sulcus non-small cell lung Shaw RR, Paulson DL, Kee JL Jr (1961) Treatment of the
cancers: report of Japan Clinical Oncology Group Trial superior sulcus tumor by irradiation followed by resection.
9806. J Clin Oncol 26:644–649 Ann Surg 154:29–40
Mansour Z, Kochetkova EA, Santelmo N et al (2008) Persistent Shen KR, Meyers BF, Larner JM, Jones DR (2007) Special
N2 disease after induction therapy does not jeopardize early treatment issues in lung cancer: ACCP guidelines evidence-
and medium term outcomes of pneumonectomy. Ann base clinical practice guidelines (2nd edition). Chest
Thorac Surg 86:228–233 132:290S–305S
Marra A, Eberhardt W, Pöttgen C et al (2007) Induction Thomas M, Rübe C, Hoffknecht P et al (2008) Effect of
chemotherapy, concurrent chemoradiation and surgery for preoperative chemoradiation in addition to preoperative
pancoast tumour. Eur Resp J 29:117–127 chemotherapy: a randomized trial in stage III non-small cell
Misthos P, Papagiannakis G, Kokotsakis J et al (2007) Surgical lung cancer. Lancet Oncol 9:636–648
management of lung cancer invading the aorta or the Trousse D, D’Journo XD, Avaro JP (2008) Multifocal T4 non-
superior vena cava. Lung Cancer 56:223–227 small cell lung cancer: a subset with improved prognosis.
Molina JR, Yang P, Cassivi SD et al (2008) Non-small cell lung Eur J Cardiothorac Surg 33:99–103
cancer: epidemiology, risk factors, treatment and survivor- Uy KL, Darling GD, Xu W et al (2007) Improved results of
ship. Mayo Clin Proc 83:584–594 induction chemoradiation before surgical intervention for
Mountain CF (1997) Revisions in the international system for selected patients with stage IIIA-N2 non-small cell lung
staging lung cancer. Chest 111:1710–1717 cancer. J Thorac Cardiovasc Surg 134:188–193
NCCN clinical practice guidelines in oncology. Non-small cell Van Meerbeeck JP, Kramer GW, Van Schil PE et al (2007)
lung cancer.V.2.2009. www.nccn.org Randomized controlled trial of resection versus radio-
Port JL, Korst RJ, Lee PC et al (2007) Surgical resection for therapy after induction chemotherapy in stage IIIA-N2
multifocal (T4) non-small cell lung cancer: is the T4 non-small cell lung cancer. J Natl Cancer Inst 99:
designation valid? Ann Thorac Surg 83:397–401 442–450
Pourel N, Santelmo N, Naafa N et al (2008) Concurrent Van Schil P, Van Meerbeeck J, Kramer G et al (2005)
cisplatin/etoposide plus 3D-conformal radiotherapy fol- Morbidity and mortality in the surgery arm of EORTC
lowed by surgery for stage IIB (superior sulcus T3N0)/III 08941 trial. Eur Resp J 26:192–197
non-small cell lung cancer yields a high rate of pathological Wu L, Xu Z, Zhao X, et al. (2009) Surgical treatment of lung
complete response. Eur J Cardiothorac Surg 13:829–836 cancer invading the left atrium of base of the pulmonary
Preoperative chemoradiotherapy vs. chemotherapy alone in vein. World J Surg (published online DOI: 10.1007/
non-small cell lung cancer (NSCLC) patients with medias- s00268-008-9873-5)
tinal lymph node metastases (Stage IIIA, N2): a randomized Yap S-P, Lim W-T, Foo K-F et al (2008) Induction concurrent
prospective Phase III trial. NCT00030771. Bethesda MD: chemoradotherapy using paclitaxel and carboplatin combi-
National Library of Medicine, 2009. (Accessed Feb 5, 2011 nation followed by surgery in locoregionally advanced
at http://www.clinicaltrials.gov) non-small cell lung cancer—asian experience. Ann Acad
Rao J, Sayeed RA, Tomaszek S et al (2007) Prognostic factors Med Singapore 37:377–382
in resected satellite-nodule T4 non-small cell lung cancer. Yildizeli B, Dartevelle PG, Fadel E et al (2008) Results of
Ann Thorac Surg 84:934–939 primary surgery with T4 non-small cell lung cancer during a
Robinson LA, Ruckdeschel JC, Wagner H et al (2007) 25-year period in a single centre: the benefit is worth the
Treatment of non-small cell lung cancer stage IIIA: ACCP risk. Ann Thorac Surg 86:1065–1075
Prophylactic Cranial Irradiation
Jason Francis Lester

Contents Abstract
The incidence of brain metastases following loco-
1 Introduction.............................................................. 445 regional treatment for locally advanced non-small
2 Risk Factors for Brain Metastases cell lung cancer (LA-NSCLC) is high. Brain
Development ............................................................. 446 metastases impair quality of life and are associated
2.1 Disease Stage............................................................. 446 with a poor prognosis. The rationale behind
2.2 Histological Subtype ................................................. 446
prophylactic cranial irradiation (PCI) is to control
2.3 Age ............................................................................. 446
2.4 Neoadjuvant Chemotherapy ...................................... 447 or eradicate undetectable micrometastases before
they become clinically significant without inducing
3 Randomized Controlled Trials of PCI
in NSCLC ................................................................. 447 severe adverse effects. Given the profound detri-
3.1 VALG Trial ............................................................... 447 mental effect on survival, treatment that reduces
3.2 Umsawasdi Trial........................................................ 447 the incidence of brain metastases in patients with
3.3 RTOG 84-03 .............................................................. 448 LA-NSCLC might reasonably be expected to
3.4 SWOG Trial............................................................... 448
3.5 RTOG 0214 ............................................................... 448 prolong life and improve quality of life. This
chapter reviews the role of PCI in LA-NSCLC
4 Incidence of Brain Metastases ............................... 448
with specific reference to the five randomized
5 Time to Brain Metastases....................................... 449 controlled trials carried out in this field.
6 Survival ..................................................................... 449
7 Toxicity...................................................................... 450
8 Quality of Life.......................................................... 450 1 Introduction
9 Neurocognitive Function......................................... 450
The incidence of brain metastases following locore-
10 What is the Most Effective PCI Regimen? .......... 451 gional treatment for locally advanced non-small cell
11 Summary................................................................... 451 lung cancer (LA-NSCLC) is high; in up to 30% of
References.......................................................................... 451 cases, the brain is the first site of relapse (Stuschke
et al. 1999). In addition, studies have shown that brain
metastases will occur in up to half of patients during
the course of their disease (Strauss et al. 2005; Law
et al. 1997). Brain metastases can result in debilitating
symptoms, significantly impair quality of life (QOL)
and dramatically shorten life expectancy (Nussbaum
et al. 1996; Gaspar L et al. 1997).
J. F. Lester (&) A study comparing two cranial radiotherapy (RT)
Velindre Cancer Centre, Cardiff, UK
e-mail: jason.lester@velindre-tr.wales.nhs.uk
regimens in patients with solid tumors starkly illustrates

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 445
DOI: 10.1007/174_2011_252, Ó Springer-Verlag Berlin Heidelberg 2011
446 J. F. Lester

the poor prognosis of NSCLC patients with brain 2.1 Disease Stage
metastases; the subgroup of patients in the trial with
NSCLC had a median survival of just 69 days (Priest- The incidence of brain metastases depends on the
man et al. 1996). initial disease stage. Salbeck et al. (1990) reported
Adjuvant chemotherapy has been shown to prolong no cases of brain metastases on initial CT staging in
survival after potentially curative resection for NSCLC patients with stage I and II NSCLC. In the same
(Arriagada et al. 2010). It is unlikely, however, that study, CT scanning detected brain metastases in
adjuvant chemotherapy decreases the incidence of brain 17.5% of patients thought to have stage III disease.
metastases as the normal brain is considered a sanctuary The rate of brain metastases as a first site of relapse
site for chemotherapy. This is supported by the findings of in stage III disease is reported to be as high as 30%
the international adjuvant lung cancer trial (IALT) at four years (Stuschke et al. 1999). Indeed, there is
(Dunant et al. 2005). IALT was designed to assess the some evidence that patients with stage IIIB disease
potential benefit of adjuvant cisplatin-based chemother- may have a higher incidence of brain metastases
apy after complete resection of NSCLC. The incidence of than those with stage IIIA disease (Robnett et al.
local recurrence and distant metastases was better in the 2001). Therefore, it would seem PCI should be more
chemotherapy arm, but there was no reduction in the beneficial in stage III as opposed to early stage
incidence of brain metastases; brain as the first site of disease.
metastases was seen in 18.1% of patients in the adjuvant
chemotherapy arm compared to 16.3% in the control arm.
In small cell lung cancer (SCLC), prophylactic 2.2 Histological Subtype
cranial irradiation (PCI) has been shown to signifi-
cantly reduce the incidence of brain metastases and Studies have suggested brain metastases occur in a
prolong survival. For SCLC patients treated with higher proportion of patients with adenocarcinoma or
curative intent achieving complete or near-complete large cell carcinoma (Figlin et al. 1988; Perez et al
remission with chemotherapy, survival is increased by 1987; Salbeck et al. 1990). For example, Perez et al.
5.4% at three years and the cumulative rate of brain (1987) reported on patterns of failure in 551 patients
metastases reduced by 25.3% (PCIOCG 2002). with inoperable NSCLC treated with definitive RT.
Despite the high incidence of brain metastases in The brain was the initial site of relapse in 7%
patients with LA-NSCLC, the role of PCI has not of patients with squamous carcinoma, 13% with
been established (Lester et al. 2005). large cell carcinoma and 19% with adenocarcinoma.
There have been five published randomized con- During the entire course of the disease, 16% of
trolled trials (RCTs) of PCI versus no PCI in patients squamous patients and 30% of adenocarcinoma and
with LA-NSCLC following locoregional treatment large cell carcinoma patients developed brain metas-
(Cox et al. 1981; Umsawasdi et al. 1984; Russell tases. It would be expected that any benefit from PCI
et al. 1991; Miller et al. 1998; Gore et al. 2011). would be more pronounced in patients with the latter
This chapter will review the literature on PCI for two histological subtypes.
LA-NSCLC with specific reference to these trials and
other relevant publications.
2.3 Age

2 Risk Factors for Brain Metastases There is some evidence that younger patients may be
Development at higher risk of developing brain metastases as the
first site of failure. Carolan et al. (2005) reported on
The risk of developing brain metastases after loco- patients with LA-NSCLC treated with curative intent.
regional treatment for LA-NSCLC depends on several Patients aged less than 60 had a 25.6% risk of brain
factors. In order to minimize the number of patients metastases compared to an 11.4% risk for patients
treated unnecessarily, it would be beneficial to iden- aged 60 and over. This trend has been reported by
tify a high risk population which might derive more several other authors (Ceresoli et al. 2002; Westeel
benefit from PCI. 2003).
Prophylactic Cranial Irradiation 447

Table 1 Published RCTs of PCI in LA-NSCLC


RCT No. Thoracic PCI Brain met Survival
treatment dose incidence (PCI vs no)
(PCI vs no)
VALG 410 RT: 50 Gy/25F/5 weeks or 42 Gy/15F/3 weeks 20 Gy/10F 6 vs 13% 35.4 vs
(S) 41.4 weeks
(NS)1
Umsawasdi 100 Chemo-RT (50 Gy/25F/5 weeks), or 30 Gy/10F 4 vs 27% 22 vs 23.5% at
combinations of surgery, RT and chemo (S) 3 years
RTOG 84-03 187 RT: 55–60 Gy/30F/6 weeks Post op: 50 Gy/ 30 Gy/10F 9 vs 19% 8.4 vs
25F/5 weeks (NS) 8.1 months
(NS)1
SWOG 254 Primary RT: 58 Gy/29F or Neoadjuvant chemo 37.5 Gy/15F Or 1 vs 11% 8 vs 11 months
then RT and adjuvant chemo 30 Gy/15F (S) (S)1
RTOG 0214 356 RT alone ([30 Gy) or combinations of surgery, 30 Gy/15F 7.7 vs 18% 75.6 vs 76.9%
RT and chemo (S) (S) at 1 year
No number of patients, RT radiotherapy, PCI prophylactic cranial irradiation, F fractions, chemo chemotherapy, S significant,
NS not significant
1
Median survival

2.4 Neoadjuvant Chemotherapy widely in the local therapy used and PCI dose-
fractionation schedules. All five trials required histo-
Some authors have suggested that the risk is higher logical confirmation of the diagnosis. In four trials,
when chemotherapy is given prior to definitive local patients were randomized to PCI or observation irre-
therapy. Andre et al. (2001) reported on relapse pat- spective of thoracic response to treatment; in RTOG
terns in 109 patients with N2 disease treated with 0214, patients needed to have non-progressive disease
neoadjuvant chemotherapy prior to surgery compared after local treatment (Gore et al. 2011). The trial
to 185 patients treated with primary surgical resec- designs are outlined below:
tion. Brain metastases occurred in 22% of patients
given neoadjuvant chemotherapy and 11% having
primary surgery. 3.1 VALG Trial
One possible explanation for this is that neoadjuvant
chemotherapy delays definitive local therapy and The VALG trial randomized 410 evaluable male
allows more time for the primary tumor to metastasize. patients not considered suitable for surgical resection
There are factors then, which are associated with with no spread beyond the regional nodes to one of two
an increased risk of brain metastases. The strongest radical RT regimens; 50 Gy in 25 fractions over 5 weeks
association is with disease stage, and all five pub- or 42 Gy in 15 fractions over 3 weeks (Cox et al. 1981).
lished RCTs of PCI in NSCLC mainly or exclusively Overall, 323 patients were evaluable. In the PCI arm,
included patients with locally advanced disease. patients received 20 Gy in 10 fractions over 2 weeks.

3 Randomized Controlled Trials 3.2 Umsawasdi Trial


of PCI in NSCLC
One hundred patients with LA-NSCLC of any cell
There have been five published RCTs of PCI versus type were randomized and 97 patients were evaluable
no PCI in patients with LA-NSCLC following loco- (Umsawasdi et al. 1984). Of these, 87% were stage III
regional treatment which are summarized in Table 1 and 13% were stage I to II. The thoracic treatment
(Cox et al. 1981; Umsawasdi et al. 1984; Russell et al. received was not clearly stated for all patients. Sixty-
1991; Miller et al. 1998; Gore et al. 2011). The five three patients received radical chemo-RT (thoracic
studies included different patient groups, and varied RT dose 50 Gy in 25 fractions over 5 weeks) and
448 J. F. Lester

34 patients received differing combinations of sur- et al. 1998; Gore et al. 2011). The results from RTOG
gery, RT and chemotherapy. PCI patients received 8403 although not statistically significant, also
30 Gy in 10 fractions over 2 weeks. strongly support the effectiveness of PCI in reducing
the incidence of brain metastases.
In the VALG study, the two thoracic RT schedules
3.3 RTOG 84-03
used were combined for statistical analysis (Cox et al
1981). The incidence of brain metastases was signif-
RTOG 84-03 randomized 187 patients with inopera-
icantly lower in the PCI arm compared to the obser-
ble or unresectable adenocarcinoma or large cell
vation arm (6 vs 13%, P = 0.038). Subgroup analysis
carcinoma confined to the chest and resected carci-
showed the only specific cell type in which PCI was
nomas of the same cell types (Russell et al 1991).
significantly more effective in reducing the incidence
Patients received primary RT 55–60 Gy in 30 frac-
of brain metastases was adenocarcinoma (0 vs 29%,
tions over 6 weeks or 50 Gy in 25 fractions over
P = 0.04).
5 weeks to the mediastinum and hilar areas following
It might be expected that any benefit from PCI is
surgical resection. PCI patients received 30 Gy in 10
more likely to be seen in patients with adenocarci-
fractions over 2 weeks.
noma, as these patients have a higher incidence of
brain metastases (Perez et al. 1987).
3.4 SWOG Trial In Umsawasdi et al. (1984) the incidence of brain
metastases in the PCI arm was 4% compared to 27%
The SWOG trial randomized 254 patients with inop- in the observation arm (P = 0.02). Multivariate
erable stage III NSCLC (Miller et al. 1998). Patients analysis suggested the beneficial effect of PCI was
were first randomized to either chest RT (58 Gy in 29 only significant in females, patients with a good
fractions) or neoadjuvant chemotherapy followed by performance status, weight loss less than 6%, squa-
chest RT and adjuvant chemotherapy. The first 34 mous histology and stage III disease. The benefit for
patients in the PCI arm received 37.5 Gy in 15 frac- squamous cancers only is counter-intuitive as brain
tions, but this was changed to 30 Gy in 15 fractions metastases are more common in patients with ade-
soon after the trial began recruiting due to concerns nocarcinoma (Perez et al. 1987). Only 97 patients in
about early deaths in the PCI arm. total were evaluable, so sample sizes in the subgroup
analysis may have been too small to reliably estimate
differences.
3.5 RTOG 0214 In the SWOG trial the incidence of brain metas-
tases in the PCI arm was 1% compared to 11% in the
RTOG 0214 randomized 356 patients with stage III observation arm (P = 0.003) (Miller et al. 1998). No
NSCLC without disease progression after surgery and/ Subgroup analysis was published.
or RT with or without chemotherapy (Gore et al. 2011). RTOG 0214 was the only trial to include regular
Local treatment was extremely variable; patients could brain imaging as part of the follow-up protocol (Gore
have RT alone ([30 Gy) or RT with neoadjuvant, et al. 2011) CT head was required at 6 and 12 months
concurrent or adjuvant chemotherapy, surgery alone or then yearly after RT. The incidence of brain metas-
surgery with pre- or post-operative chemotherapy, RT tases was lower in the PCI arm compared to the
or both. RTOG 0214 had planned to recruit 1058 control arm at 6 months (3.3 vs 10.7%, P = 0.004)
patients but closed early due to poor recruitment. The and 1 year (7.7 vs 18%, P = 0.004). No subgroup
PCI schedule used was 30 Gy in 15 fractions. analysis was published.
In RTOG 84-03, PCI did not significantly reduce
the incidence of brain metastases compared to the
4 Incidence of Brain Metastases observation arm (9 vs 19%, P = 0.10) (Russell et al.
1991). The prevalence of brain metastases at
PCI significantly reduced the incidence of brain 24 months for PCI was 15 versus 31% in the obser-
metastases in four of the five published randomized vation arm. Again, this result was not statistically
trials (Cox et al. 1981; Umsawasdi et al. 1984; Miller significant but RTOG 84-03 results strongly suggest
Prophylactic Cranial Irradiation 449

PCI reduces the incidence of brain metastases and 34 patients in the PCI arm received 37.5 Gy in 15
that this benefit is maintained two years after PCI. fractions, but this was changed to 30 Gy in 15 frac-
Looking at the results of all five RCTs together, it tions soon after the trial began recruiting due to
is clear that PCI is effective at reducing the incidence concerns about early deaths in the PCI arm. The
of brain metastases in patients with LA-NSCLC given remaining patients randomized to PCI received 30 Gy
locoregional treatment for their disease. Attempts to in 15 fractions, but had a similar median survival to
define particular subgroups which may derive pro- the 37.5 Gy group. It was not clear to the investiga-
portionally more benefit from PCI have been largely tors why there was a shorter life expectancy in the
unsuccessful due to the relatively small size of the PCI arm, but PCI was given concurrently with
trials carried out. thoracic RT and it may be that the subsequently
increased toxicity contributed to the reduced survival.
The only RCT to mandate patients needed to
5 Time to Brain Metastases have at least stable disease after locoregional treat-
ment was RTOG 0214 (Gore et al. 2011). It is
In the VALG trial, the median time to development of reasonable to assume that any survival advantage
brain metastases was 34 weeks in the PCI group and with PCI is far more likely to be seen in patients
29 weeks in the observation group (Cox et al. 1981). with controlled thoracic disease. Patients who have
The statistical significance of this result was not progressive or metastatic disease after locoregional
reported, but the difference is clearly not a large one. In treatment will have a short life expectancy and are
Umsawasdi et al. (1984), PCI significantly prolonged likely to die from extracranial disease complications
the median time to brain metastases (50.5 vs 23 weeks, before any benefit from PCI can be seen. Despite
P = 0.002). The SWOG, RTOG 84-03 and RTOG this, there was no difference in 1-year overall sur-
0214 trials did not report on time to brain metastases. vival in RTOG 0214 (75.6% for PCI vs 76.9% for
The only RCT to show a significant benefit was the controls). The lack of survival advantage in RTOG
smallest of the 5 trials, so it remains unclear as to 0214 and the other RCTs is not necessarily sur-
whether PCI delays time to brain metastases. prising. RTOG 0214 initially planned to randomize
1058 patients; this was the number of patients
needed to detect a 20% difference in overall survival
6 Survival with 80% statistical power. The trial closed early
due to poor recruitment with only 340 evaluable
To date, no trial has reported a survival advantage with patients enrolled, so any true benefit is unlikely to
PCI over observation despite impressive reductions in have been seen. Applying the same statistical logic
the incidence of brain metastases in all published to the other four RCTs, it is clear that none of the
RCTS. The median survival figures for PCI versus studies was large enough to detect a survival benefit
observation in the VALG trial were 35.4 weeks from PCI if one truly exists - the largest study was
versus 41.4 weeks (P = 0.5), and in RTOG 84-03, the VALG study and this only included 410 evalu-
8.4 months versus 8.1 months (P = 0.36) (Cox et al. able patients.
1981; Russell et al. 1991). RTOG 84-03 also reported The experience with PCI in SCLC is also
no significant difference between PCI and observation important to consider. Several trials suggested a
in one and two-year survival (40 vs 44% and 13 vs 21%, reduction in the incidence of brain metastases with
P = 0.36) (Russell et al. 1991). PCI, but no convincing improvement in survival.
In Umsawasdi et al. (1984), three-year survival in As a consequence, PCI was not considered a stan-
the PCI and control groups was 22 and 23.5%, dard of care in SCLC. A subsequent meta-analysis
respectively. No statistical analysis of the survival of seven RCTs including 987 patients did however,
data was reported but it is highly unlikely there was a demonstrate a small but significant increase in three-
real difference between the arms. year survival of 5.4% with PCI (PCIOCG 2002). It
In the SWOG trial, median survival was actually is not logical to extrapolate these results to NSCLC
significantly shorter in the PCI arm (8 vs 11 months, patients as SCLC is a more radiosensitive disease
P = 0.004) (Miller et al. 1998). In this study, the first with a higher incidence of brain metastases, but it
450 J. F. Lester

may be that a large enough RCT would demonstrate It is difficult to draw any definitive conclusions
a survival advantage not seen in the relatively small regarding PCI-related toxicity due to the relative
trials reported to date. paucity of data. It does seem that from the RTOG
In addition, the locoregional treatment for many 0214 results, PCI at a dose of 30 Gy in 15 fractions is
patients in the five RCTs would not be considered generally well tolerated.
optimal by today’s standards. For example, even in
RTOG 0214, the most contemporary of the trials,
patients were eligible having had as little as 30 Gy RT 8 Quality of Life
to the chest. This is not a tumoricidal RT dose and is
unlikely to achieve long–term local disease control. Prospective QOL assessments were carried out in
Patients would then be at risk of disease relapse and RTOG 0214—none of the other randomized trials
early death before any PCI benefit could be seen. The included QOL data collection. QOL was assessed
disease-free survival at 1 year was not significantly using the EORTC QLQ-C30 and BN20 question-
different between the two arms in RTOG 0214 despite naires. In total, 340 patients were evaluable in RTOG
a significant decrease in the incidence of brain 0214. At baseline, 95% of patients completed QOL
metastases with PCI suggesting patients are relapsing assessments but compliance to testing declined rap-
systemically and dying, possibly before any PCI idly during the study. At 3 months, only 43% of
benefit can be seen. potentially evaluable patients completed the assess-
Summarizing then, no RCT had shown a survival ments and this fell further to 34% at 12 months. There
benefit from PCI in LA-NSCLC despite all showing a were no significant differences between the two arms
reduction in the incidence of brain metastases. The in change in QOL scores from baseline at 6 and
reasons for this may in part be down to trial design; 12 months. In the trial, subgroup analysis was carried
individual trials had relatively few participants, out to try and identify patients at higher risk of
included some patients with early stage disease, and experiencing a decline in QOL. No clear differences
locoregional treatment was suboptimal and varied. emerged in any of the subgroups tested.
Two trials have evaluated the effect of PCI on QOL
in SCLC patients (Arriagada et al. 1995; Gregor et al.
7 Toxicity 1997). Neither study reported any difference in QOL
between patients in the PCI arm and control arm.
Toxicity data collection and subsequent publication Given the relatively small number of patients
was poor in four of the five trials. Only RTOG 0214 evaluated in RTOG 0214 and the lack of data from
included detailed prospective toxicity data collection other trials, definitive conclusions cannot be drawn on
(Gore et al. 2011). The VALG trial did not report on the effect of PCI on QOL. It does seem that any effect
PCI-related toxicity (Cox et al. 1981). RTOG 84-03 on QOL is unlikely to be substantial within the first
reported no acute toxicity other than epilation and 12 months, but confirmatory research is needed and
skin reactions and no late toxicity (Russell et al. longer follow-up data is essential.
1991). The Umsawasdi and SWOG trials reported no
excessive toxicity with PCI compared to the obser-
vation arm, but it was not stated in either trial what 9 Neurocognitive Function
data was collected or how this data was collected
(Umsawasdi et al. 1984; Miller et al, 1998). RTOG 0214 was the only RCT to collect detailed
RTOG 0214 reported that PCI resulted in generally prospective data on the long-term effects of PCI on
mild toxicity (Gore et al. 2011). Grade 3 and 4 tox- neurocognitive function (NCF) (Gore et al. 2011).
icities occurred in 4 and 1% of patients, respectively. NCF was assessed using the Mini-Mental Status
The grade 3 toxicities reported were acute fatigue, Examination (MMSE), Hopkins Verbal Learning Test
dyspnoea, ataxia, and depression. One patient repor- (HVLT) and Activity of Daily Living Scale (ADLS).
ted acute grade 4 mood alteration/depression. Four The MMSE change scores from baseline at
patients reported grade 3 late toxicity including 3 months showed significantly more patients reported
dyspnoea, syncope, weakness, and fatigue. a decline in the PCI arm than in the control arm (36 vs
Prophylactic Cranial Irradiation 451

21%, P = 0.04). A similar trend was seen in the


HVLT, with significantly more patients in the PCI 10 What is the Most Effective PCI
arm reporting deterioration in immediate recall (45 vs Regimen?
13%, P \ 0.001) and delayed recall (44 vs 10%,
P \0.001). At the 6 month time point, the differences Four different PCI regimens were used in the five
in both MMSE and HVLT had disappeared. Intrigu- RCTs (Table 1), but the small number of patients in
ingly, at the 12 month time point there remained no each trial, differences in inclusion criteria and loco-
difference in the MMSE scores, but immediate recall regional treatment make any comparison between the
(26 vs 7%, P = 0.03) and delayed recall (32 vs 5%, trials inappropriate. In addition, no randomized trial
P = 0.008) in the HVLT were again significantly has compared these (or any other) PCI regimens head-
worse in the PCI arm. to-head. Therefore, it is not possible to establish
It is likely that the decline in recall is a conse- which PCI regimen is superior.
quence of PCI; the differences seen in the tests may be
because HVLT has a better sensitivity than the
MMSE for detecting early dementia (Wade 1992). 11 Summary
What is not known is whether this decline stabilizes
or continues to deteriorate. Longer term NCF data There is insufficient evidence to support the routine
from the RTOG 0214 may become available in future use of PCI in the management of patients with
to help answer this question. LA-NSCLC having undergone locoregional treat-
It is worthwhile considering the data on PCI in ment. All five published RCTs show a reduction in the
SCLC when attempting to define the effect of PCI on incidence of brain metastases in patients receiving
NCF. Two trials have evaluated the effect of PCI PCI, but this did not translate into a survival advan-
on NCF in SCLC patients. In Arriagada et al. (1995) tage. The patient population most likely to benefit is
patients in the PCI group received 24 Gy in 8 frac- not clear, nor is the most beneficial RT schedule.
tions over 12 days. There were no significant differ- Unlike in SCLC, the trials are too heterogenous for
ences between patients receiving PCI and those in the meta-analysis to be useful, and the poor recruitment
observation group in terms of NCF. In Gregor et al. to the RTOG 0214 trial means the role of PCI in
(1997) the majority of patients randomized to PCI LA-NSCLC may never fully be established.
received 30 Gy in 10 fractions over two weeks.
In both groups, there was an impairment of NCF before
PCI, and further impairment at 6 and 12 months, but no References
additional impairment in the PCI group compared to the
control group. It is reasonable to assume that any effect Andre F, Grunenwald D, Pujol JL et al (2001) Patterns of
on NCF with a given PCI regimen would be similar in relapse of N2 non-small cell lung carcinoma patients treated
with preoperative chemotherapy: should prophylactic cra-
SCLC and NSCLC patients. nial irradiation be reconsidered? Cancer 91:2394–2400
It is likely, therefore, that there is an acute Arriagada R, Le Chevalier T, Borie F (1995) Prophylactic
reversible decline in NCF after PCI which improves cranial irradiation for patients with small-cell lung cancer in
by 6 months. Longer term follow-up data is lacking, complete remission. J Nat Cancer Inst 87:183–190
Arriagada R, Auperin A et al (2010) Adjuvant chemotherapy,
but it is probable that deterioration in NCF can occur with or without postoperative radiotherapy, in operable non-
12 months after PCI. It is not yet known whether this small-cell lung cancer: two meta analyses of individual
decline is progressive. patient data. Lancet 375:1267–1277
RT techniques are being developed that may Carolan H, Sun AY, Bezjak A et al (2005) Does the incidence
and outcome of brain metastases in locally advanced non-
reduce the late cognitive sequelae of treatment. The small cell lung cancer justify prophylactic cranial irradiation
Hippocampus is a structure that is crucial to NCF and or early detection? Lung Cancer 49:109–115
hippocampal metastases are rare. Sparing of this Ceresoli GL, Reni M, Chiesa G et al (2002) Brain metastases
region with PCI is feasible using advanced RT plan- in locally advanced non-small cell lung carcinoma after
multimodality treatment: risk factors analysis. Cancer 95:
ning and delivery systems (Marsh et al. 2010, Hsu 605–612
et al. 2010). The hope is that these technical advances Cox JD, Stanley K, Petrovich Z et al (1981) Cranial Irradiation in
translate into meaningful benefits for patients. Cancer of the Lung of All Cell Types. JAMA 245:469–472
452 J. F. Lester

Dunant A, Pignon J-P, Le Chevalier T (2005) Adjuvant PCIOCG (The Prophylactic Cranial Irradiation Overview Col-
Chemotherapy for Non–Small Cell Lung Cancer: Contri- laborative Group). Cranial irradiation for preventing brain
bution of the International Adjuvant Lung Trial. Clin metastases of small cell lung cancer in patients in complete
Cancer Res 11:5017s remission. Cochrane database of systematic reviews (2002),
Figlin RA, Piantadosi S, Field R (1988) Intracranial recurrence Issue 4. doi:10.1002/14651858.CD002805
of carcinoma after complete surgical resection of stage I, II Perez CA, Pajak TF, Rubin P et al (1987) Long term
and III non-small cell lung cancer. N Engl J Med 318:1300– observations of the patterns of failure in patients with
1305 unresectable non-oat cell carcinoma of the lung treated with
Gaspar L, Scott C et al (1997) Recursive partitioning analysis definitive radiotherapy. Cancer 59:1874–1881
(RPA) of prognostic factors in three radiation therapy Priestman TJ, Dunn J et al (1996) Final results of the RCR
oncology group (RTOG) brain metastases trials. Int J Rad trial comparing two different radiotherapy schedules in
Onc Biol Phys 37:745–751 the treatment of cerebral metastases. Clin Oncol 8:308–
Gore EM, Bae K, Wong SJ et al (2011) Phase III comparison of 315
prophylactic cranial irradiation versus observation in Robnett TJ, Machtay M, Stevenson JP et al (2001) Factors
patients with locally advanced non-small-cell lung cancer: affecting the risk of brain metastases after definitive
primary analysis of radiotherapy oncology group study chemoradiation for locally advanced non-small cell lung
RTOG 0214. J Clin Oncol 29:272–278 carcinoma. J Clin Oncol 19:1344–1349
Gregor A, Cull A, Stephens RJ et al (1997) Prophylactic cranial Russell AH, Pajak TE, Selim HM et al (1991) Prophylactic
irradiation is indicated following complete response to cranial irradiation for lung cancer patients at high risk for
induction therapy in small cell lung cancer: results of a development of cerebral metastasis: results of a prospective
multicentre randomised trial. Eur J Cancer 33:1752–1758 randomised trial conduced by the radiation therapy oncol-
Hsu F, Carolan H, Nichol A et al (2010) Whole brain ogy group (RTOG 84–03). Int J Radiat Oncol Biol Phys
radiotherapy with hippocampal avoidance and simultaneous 21:637–643
integrated boost for 1–3 brain metastases: a feasibility study Salbeck R, Grau HC, Artmann H (1990) Cerebral tumor staging
using volumetric modulated arc therapy. Int J Radiat Oncol in patients with bronchial carcinoma by computed tomog-
Biol Phys 76:1480–1485 raphy. Cancer 66:2007–2011
Law A, Daly B, Madsen M et al (1997) High incidence of Strauss GM, Herndon JE, Sherman DD et al (2005) Neoadju-
isolated brain metastases following complete response in vant chemotherapy and radiotherapy followed by surgery in
advanced non-small cell lung cancer: a new challenge. Lung stage III non-small cell carcinoma of the lung: a report of a
Cancer 18:65s cancer and leukemia group B phase II study. J Clin Oncol
Lester JF, Macbeth FR, Coles B (2005) Prophylactic cranial 10:1237–1244
irradiation for preventing brain metastases in patients Stuschke W, Eberhardt W, Pottgen C et al (1999) Prophylactic
undergoing radical treatment for non-small cell lung cancer: cranial irradiation in locally advanced non-small-cell lung
a cochrane review. Int J Radiat Oncol Biol Phys 63:690–694 cancer after multimodality treatment: long term follow-up
Marsh JC, Godbole RH, Herskovic AM et al (2010) Sparing of and investigations of late neuropsychologic effects. J Clin
the neural stem cell compartment during whole brain Oncol 17:2700–2709
radiation therapy: a dosimetric study using helical tomo- Umsawasdi T, Valdivieso M, Chen TT et al (1984) Role of
therapy. Int J Radiat Oncol Biol Phys 78:946–954 elective brain irradiation during combined chemoradiother-
Miller TP, Crowley JJ, Mira J et al (1998) A randomized trial of apy for limited disease non-small cell lung cancer. J Neuro-
chemotherapy and radiotherapy for stage III non-small cell Oncol 2:253–259
lung cancer. Canc Ther 4:229–236 Wade DT (1992) Measurement in neurological rehabilitation.
Nussbaum ES, Djalilian HR, Cho KH et al (1996) Brain Oxford University Press, NY
metastases: histology, multiplicity, surgery, and survival. Westeel V (2003) Risk of brain metastases in non-metastatic
Cancer 78:1781–1788 non-small cell lung cancer. Lung Cancer 41:21s
Palliative External Beam Thoracic Radiation
Therapy of Non-Small Cell Lung Cancer
Stein Sundstrøm

Contents Abstract
About two-thirds of patients with non-small cell
1 Introduction.............................................................. 454 lung cancer are diagnosed with incurable disease
2 Symptoms.................................................................. 454 and are usually treated with a palliative intent.
Palliative radiotherapy is defined as radiotherapy
3 Palliative Radiotherapy: Definition ....................... 454
given with less than radical doses. Although a
4 Prognostic Factors for Selecting Treatment ........ 454 large variation in treatment schedules considering
5 Radiotherapy Technique......................................... 455 dose, fractionation, and overall treatment time are
6 Defining the Radiotherapy Volume ....................... 455
used, usually a dose 50 Gy is considered as
palliative. Given the palliative intent of the radio-
7 Dose and Fractionation ........................................... 455
therapy with the goal of reducing tumour-related
8 Phase III Studies...................................................... 456 symptoms, the radiotherapy should be simple to set
9 Interpretation ........................................................... 457 up, to perform, and less time-consuming for the
patient. If high-dose palliative fractionated radio-
10 Treatment Recommendations ................................ 457
therapy is planned a normal setup margin defining
11 When Should Palliative Radiotherapy be GTV and CTV/PTV is recommended including the
Delivered? ................................................................. 458
tumour and disease-related nodes. When low-dose
12 Summary................................................................... 458 radiotherapy is planned, the treated volume should
References.......................................................................... 458 include the symptomatic part of the tumour.Most
studies show that the effect on symptoms and
palliative effect is similar regardless of dose and
fractionation. A trend of more rapid relief of
symptoms in favour of hypofractionation is
observed. Though no major difference in median
survival is observed, some patients with localised
stage III disease may have better survival with a
protracted high-dose schedule. Acute toxicity with
dysphagia is mild, temporary, and manageable.
Late toxicity is rare and sporadic with low severity.
Palliative thoracic radiotherapy should not be
administrated to patients without symptoms
present.
S. Sundstrøm (&)
St Olav University Hospital, Trondheim, Norway
e-mail: Stein.Sundstrom@stolav.no

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 453
DOI: 10.1007/174_2011_316,  Springer-Verlag Berlin Heidelberg 2011
454 S. Sundstrøm

1 Introduction 4 Prognostic Factors for Selecting


Treatment
The majority of patients with non-small lung cancer
are diagnosed with advanced disease, either localised Stage of the disease, spontaneous weight loss over the
stage III or metastatic stage IV disease. Approxi- last 3–6 months, and performance status are the most
mately 40% have stage III disease and 40% have robust prognostic factors in non-small cell lung can-
stage IV disease. Of stage III localised disease, only cer (Brundage et al. 2002).
one-third have tumours considered to be candidate for Disease stage is the most powerful prognosticator
a curative strategy. This leaves about two-thirds of the in lung cancer. Patients with metastatic disease have
non-small population diagnosed with incurable dis- shorter survival than stage III disease. Some good
ease and should be treated with a palliative intent. prognostic stage III patients can be treated with a
Most of these patients will have symptoms from curative intent, but even with the aim of high dose
intrathoracic tumour at diagnosis or the propensity to chemo-radiotherapy the survival is limited. Patients in
develop symptoms in the near future. In this setting, stage III with poor prognostic factors will have sur-
any intervention should have the goal of effective vival equal to stage IV disease.
palliation avoiding unacceptable toxicity. Performance status is a strong prognostic factor in
non-small cell lung cancer regardless of stage.
Patients with WHO PS status 0–1 will experience a
2 Symptoms meaningful effect of treatment, both for chemotherapy
and radiotherapy. Patients in WHO PS 2 are a bor-
The most common symptoms from tumours in the chest derline group; they will achieve less effect of che-
are dyspnoea, cough, and hemoptysis. Other symptoms motherapy, often with intolerable toxicity. Patients
are chest pain, compression of large vessels (superior with WHO PS status of 3–4 should routinely not be
vena cava syndrome), nerve infiltration causing offered chemotherapy. If treatment is required, palli-
hoarseness or Horners syndrome, and dysphagia due to ative radiotherapy is probably a better strategy for
compression of oesophagus. These latter symptoms are relieving symptoms.
related with infiltrative tumours beyond any curative Weight loss is common and is categorised
strategy. About 70% of patients with advanced disease according to the normal body weight before diagnosis
have one or more symptoms from intrathoracic tumour and to the time interval over which the weight loss
at diagnosis requiring treatment intervention (Hopwood has developed (none versus \5–10% vs. C10% of
and Stephens 1995; Lutz et al. 2001). normal body weight) over the last 3–6 months.
Weight loss is a consistent and strong prognosticator.
A weight loss C10% over the last 3 months is a strong
3 Palliative Radiotherapy: Definition negative prognostic factor, and the patient should be
treated with a palliative intent regardless of other
Palliative radiotherapy is defined as radiotherapy given prognostic factors.
with less than radical doses. Usually a dose\50 Gy is Tumour size is important regarding the possibility
considered as palliative. A large variation in treatment of eradication of a tumour by radiotherapy. There is
schedules considering dose, fractionation, and overall no general accepted consensus for the maximum
treatment time are used. The various regimes in use are tumour size or volume that precludes a radical strat-
often based on tradition, preference of the responsible egy. The pivotal study from RTOG in 1982 showed a
physician and radiotherapy capacity (Maher et al. clear relation to less effect in tumours [6 cm in
1992). The selection for technique and total dose diameter (Perez et al. 1982). Several other studies
should be based on a thorough clinical evaluation of the show the same trend (Bradley et al. 2002; Dehing-
patient before start of radiotherapy. In this setting a Oberije et al. 2008), and a tumour larger than [8–
discrimination of the patient in good prognostic or poor 10 cm is considered by most clinicians to be treated
prognostic group should be elucidated. with a palliative intent.
Palliative External Beam Thoracic Radiation Therapy of Non-Small Cell Lung Cancer 455

Table 1 Prognostic factors Good prognostic factors Poor prognostic factors


in stage III patients
WHO PS status 0–1 WHO PS status C2
Tumour size \8 cm in largest diameter Tumour size [8 cm in largest diameter
Weight loss \10% last 3 months Weight loss C10% last 3 months

Overall, in the heterogeneous stage III group of


patients, selection of either a curative or palliative 6 Defining the Radiotherapy Volume
schedule should be based on a thorough clinical
evaluation of the factors outlined in Table 1. Occur- In situations where curative intended radiotherapy is
rence of one or more of the poor prognostic factors planned there is consensus for including enlarged
should conduct a palliative treatment strategy. and disease-related lymph nodes in field planning.
Patients in stage IV disease should in almost all cases Including elective lymph nodes will add toxicity
be treated with a palliative intent. with no increase in local control or survival (Yan
et al. 2007).
If high-dose palliative fractionated radiotherapy is
5 Radiotherapy Technique planned a normal setup margin defining GTV and
CTV/PTV is recommended including the tumour and
Given the palliative intent of the radiotherapy with disease- related nodes. According to international
the goal of reducing tumour-related symptoms, the guidelines a setup margin of 1 cm from GTV to CTV,
radiotherapy should be simple to set up, to perform, and 0.5 cm from CTV to PTV is recommended.
and less time-consuming for the patient. Larger margins in the cranio-caudal direction due to
Radiotherapy planning involves consideration of respiration movements might be necessary. Reduced
patient positioning, localization of tumour, and criti- margins are desirable if the treated volume enlarge to
cal organs. A supine positioning is usually preferred [200 cm2 in field size.
with the arms adducted or above the head if oblique When low-dose radiotherapy is planned, the treated
field is planned. A conventional two-dimensional volume should include the symptomatic part of the
radiotherapy (2DRT) technique is sufficient in most tumour. In most cases this can confine the volume to the
cases based on an X-ray beam simulator. Parallel symptomatic area avoiding unnecessary radiotherapy
opposed anterior-posterior fields are used. Shielding to non-symptomatic lung tissue even though these parts
of uninvolved lung tissue should be made reducing may include tumour spread. The central airways with
side effects. If high-dose treatment is planned, oblique mediastinum and hilus on the affected side will often be
fields after maximal tolerable dose to the spine are an adequate volume. Margins from the tumour border
advocated. Previously, 40 Gy/20 fractions were con- to CTV and PTV should not be emphasized. Close
sidered as the maximal spinal cord tolerance. Recent margins or even margins set up in the tumour are
updates designate 50 Gy using conventional frac- acceptable if the volume becomes large.
tionation as safe to the thoracic spine (Kirkpatrick Considering the palliative intent of the treatment,
et al. 2010). If a more protracted schedule with higher focus on radiotherapy side effects should be kept in
palliative dose is planned, a three-dimensional con- mind. In that respect smaller volumes are more
formal radiotherapy (3DCRT) technique based on CT desirable than large volumes encompassing the
scan should be preferred. This will better define the complete tumour extension.
tumour volume and organs at risk and assure a more
precise encompassing of tumour. High energy photon
beams in the rage of 5–15 MeV is recommended. 7 Dose and Fractionation
Most radiotherapy units will in the near future
become simulator-less, basing all planning also for A large variety of fractionation schemes and radio-
palliative purposes on CT scan. This is more convenient therapy schedules are in use for palliative treatment,
for patients and time sparing for the treatment units. from single fraction of 8–10 Gy up to subradical
456 S. Sundstrøm

Table 2 Different phase III trials using low dose hypofractionated palliative TRT in advanced NSCLC
Study N Regimens Stage III/IV WHO Palliation Survival
(%) PS
MRC I (1991) 369 17 Gy/2 versus 30 Gy/10 68/32 0–2(3) Equal Equal
MRC II (1992) 235 10 Gy/1 versus 17 Gy/2 71/29 2–4 Equal Equal
MRC III (1996) 509 17 Gy/2 versus 39 Gy/13 100/0 0–2 Equal 39 Gy
better
Rees et al. (1997) 216 17 Gy/2 versus 22.5 Gy/5 Not reported 0–3 Equal Equal
Bezjak et al. (2002) 230 10 Gy/1 versus 20 Gy/5 76/24 0–3 Equal 20 Gy
better
Sundstrøm et al. (2004) 407 17 Gy/2 versus 42 Gy/15 versus 78/22 0–3 Equal Equal
50 Gy/25
Erridge et al. (2005) 148 10 Gy/1 versus 30 Gy/10 Not reported 0–3 30 Gy Equal
better
Kramer et al. (2005) 297 16 Gy/2 versus 30 Gy/10 52/48 0–3(4) 30 Gy 30 Gy
better better
Senkus-Konefka et al. 100 16 Gy/2 versus 20 Gy/5 84/16 1–3(4) Equal 16 Gy
(2005) better

doses of 50–60 Gy. These differences in treatment comparing a single fraction versus 17 Gy/2 fractions.
policy are probably due to marginal radiotherapy All trials (MRC Lung Cancer Working Party 1991,
capacity favouring low-dose irradiation. Until the first 1992, 1996; Rees et al. 1997; Bezjak et al. 2002;
study from the Medical Research Council UK was Sundstrøm et al. 2004; Erridge et al. 2005; Kramer
published in 1991 (MRC Lung Cancer Working Party et al. 2005; Senkus-Konefka et al. 2005) reported the
1991), a typical course was 30 Gy in ten fractions. effect on symptoms assessed by patients through self-
reported questionnaires and physicians’ evaluation of
symptoms, as well as overall survival. Except in two
8 Phase III Studies trials (Erridge et al. 2005; Kramer et al. 2005) where
the effect on symptoms is in favour of the higher dose,
Since the MRC I (MRC Lung Cancer Working Party the effect on disease-related symptoms are equal.
1991) study was published, eight other randomised In three trials (Medical Research Council Lung
phase III trials (Medical Research Council Lung Cancer Working Party 1996; Bezjak et al. 2002;
Cancer Working Party 1992; Medical Research Kramer et al. 2005) the survival is in favour of the
Council Lung Cancer Working Party 1996; Rees et al. high-dose arm; 39 Gy/13 fractions, 30 Gy/10 frac-
1997; Bezjak et al. 2002; Sundstrøm et al. 2004; tions, and 30 Gy/10 fractions, respectively. One trial
Erridge et al. 2005; Kramer et al. 2005; Senkus- (Senkus-Konefka et al. 2005) reports a survival ben-
Konefka et al. 2005) comparing a strict hypofrac- efit for the low-dose arm; 16 Gy/2 fractions versus
tionated schedule with a normal-fractionated regimen 20 Gy/5 fractions. In one three-armed trial
have been published, outlined in Table 2. More than (Sundstrøm et al. 2004) comparing 17 Gy/2 fractions
2,500 patients are included in these trials. All trials (n = 143) with two high-dose arms; 42 Gy/15 frac-
have either a single (8 or 10 Gy) or two large frac- tions (n = 140) and 50 Gy/25 fractions (n = 124), no
tions (17 or 16 Gy/2) as the short-course experimental difference in median survival was found.
arm. The comparative arms were fractionated sched- Five randomised phase III studies (Simpson et al.
ules with a range of 20–50 Gy. The trials included 1985; Teo et al. 1987; Abratt et al. 1995; Reinfuss et al.
patients up to WHO PS 3 with a majority of stage III 1999; Nestle et al. 2000) have compared different
patients. One trial (MRC II) (Medical Research normal to high-dose regimens, including more than
Council Lung Cancer Working Party 1992) included 1,000 patients, as shown in Table 3. Nearly all had
poor performance status patients only (WHO PS 2–4) stage III localised disease with a reasonably good
Palliative External Beam Thoracic Radiation Therapy of Non-Small Cell Lung Cancer 457

Table 3 Different phase III trials using normal-high dose palliative TRT in advanced NSCLC
Study N Regimens Stage III/ WHO Palliation Survival
IV PS
Simpson et al. 316 20 Gy/20 versus 30 Gy/10 vs 40 Gy/20 split 100/0 0–2 Equal Equal
(1985)
Teo et al. (1987) 291 31.2 Gy/4 versus 45 Gy/18 90/3 0–3 45 Gy Equal
better
Abratt et al. 84 35 Gy/10 versus 45 Gy/15 100/0 0–2 Equal Not reported
(1995)
Reinfuss et al. 240 40 Gy/10 (split 4 week) versus 50 Gy/25 vs 100/0 0–2 Not Better 40/
(1999) wait and see reported 50 Gy
Nestle et al. 152 32 Gy/16 (twice/day) versus 60 Gy/30 79/21 1–2 Equal Equal
(2000)

performance status WHO PS 0–2. Four of five studies dose schedule. As seen in Tables 2 and 3, the majority
have assessed the effect on symptoms. One has used of patients in the trials had localised stage III disease.
patient self-reported questionnaires (Nestle et al. In one trial (MRC Lung Cancer Working Party 1991)
2000), in the other studies the effect on symptoms were there was some evidence that a higher dose could give
assessed by the physicians. One study reported better some long-term survivors in stage III, even though
palliation in the high-dose arm (Teo et al. 1987). Four median survival was equal. Therefore, MRC III study
studies have data on survival, equal in three, better in (Medical Research Council Lung Cancer Working
one in the high-dose arms (Reinfuss et al. 1999). This Party 1996) was set up including only stage III dis-
study by Reinfuss et al. (1999) is special since one arm ease and good performance patients, comparing
in this three-armed trial was a ‘‘wait and see’’ arm; 17 Gy/2 fractions (F1) with high-dose 39 Gy/13
40 Gy10 (split) versus 50 Gy/25 versus ‘‘wait and fractions (F2). The median survival increased from 7
see’’. The survival in this ‘‘wait and see’’ arm was to 9 months in the F2 arm, although not significant,
inferior compared to the two actively treated arms. with a 1- and 2-year survival of 31 and 9% with 36
All studies reporting side effects show that esoph- and 12% in the F1 and F2 arms, respectively. In the
agitis and dysphagia was most frequent in the high- other study (Sundstrøm et al. 2004) comparing a strict
dose arms, although dysphagia was also detectable in low-dose with high-dose schedules, a trend in better
the hypofractionated low-dose arms. Onset and dura- survival in the high-dose arms was found. Later
tion of dysphagia was earlier and shorter in the hyp- exploration of this study restricted to stage III patients
ofractionated arms compared to high-dose arms. Rare only (Sundstrøm et al. 2006) reveal a 3- and 5-year
cases of radiation-induced myelopathy are reported, survival in the three arms (17 Gy/2, 42 Gy/15, 50 Gy/
usually mild and temporary (MacBeth et al. 1996; 25) of 1, 8, and 6%, against 0, 4, and 3%, respec-
Reinfuss et al. 2011). The incidence is estimated to 2% tively. Patients with metastatic disease can safely be
at 2-year survival for hypofractionated schedules. treated with a hypofractionated schedule.
Side effects: acute toxicity with dysphagia is mild,
temporary, and manageable. Late toxicity is rare and
9 Interpretation sporadic with low severity.

Palliation: most studies show that the effect on


symptoms and palliative effect is similar regardless of 10 Treatment Recommendations
dose and fractionation. A trend of more rapid relief of
symptoms in favour of hypofractionation is observed. Systematic reviews including a Cochrane analysis
Survival: no major difference in median survival is have been carried out focusing palliative thoracic
observed. Some patients with localised stage III dis- radiotherapy in lung cancer (Lester et al. 2006;
ease may have better survival with a protracted high- Fairchild et al. 2008). Due to large heterogeneity
458 S. Sundstrøm

concerning dose and fractionation no formal meta- progressing during or after chemotherapy with less
analysis has been initiated. The conclusion is that toxicity.
there is no substantial strong evidence that a higher Palliative thoracic radiotherapy is administered
dose gives a better outcome concerning symptom with the intention of treating symptoms from intra-
relief and survival, and that a hypofractionated regi- thoracic tumours. Patients evaluated to have less or no
men is an option for most patients. However, patients disease-related symptoms will have minimal effect of
with stage III disease with a reasonable performance immediate treatment (Falk et al. 2002; Sundstrøm
status and less weight loss should be treated with a et al. 2005). Immediate treatment is likely to give
protracted fractionated regimen 30–45 Gy. Self- unnecessary side effects like dysphagia in otherwise
reported appetite loss can help in selecting patients symptom-free patients and do not prevent the devel-
for short- or long-course palliative radiotherapy opment of later symptoms. A wait and see procedure
(Sundstrøm et al. 2006). Stage IV patients can be is therefore advocated until the patient develops dis-
treated safely with a hypofractionated technique in ease-related symptoms.
almost all cases. Some selected cases with single
metastasis can be treated more radically providing a
radical strategy for the metastatic site. Before treat- 12 Summary
ment decision is made, a thorough clinical evaluation
of the individual patient should be mandatory in all • Hypofractionated and moderately high-dose palli-
cases, elucidating the patient into a good or poor ative radiotherapy give equal effects on symptoms
prognostic group as outlined in Table 1. and survival in advanced non-small cell lung
Of special interest is the fact that palliative radio- cancer.
therapy in lung cancer can unexpectedly generate • Stage III patients not candidates for a curative
some long-term survivors (Sundstrøm et al. 2006; strategy with favourable prognostic factors should
Mac Manus et al. 2005). Approximately 1–3% of be treated with a fractionated schedule to a dose of
patients with localised disease have been found with 30–45 Gy.
5-year survival after palliative high-dose radiother- • Stage IV patients can safely be treated with a strict
apy. This can be explained by unpredictable high hypofractionated schedule.
radiotherapy sensitivity of some lung tumours. • Palliative thoracic radiotherapy should not be
administrated to patients without symptoms present.

11 When Should Palliative


Radiotherapy be Delivered? References

In the last two decades the effect of chemotherapy in Abratt RP, Shepherd LJ, Mameena Salton DG (1995) Palliative
advanced non-small cell lung cancer is recognised radiation for stage 3 non-small cell lung cancer. A
(NSCLC Meta-analysis collaborative group 2008). prospective study of two moderately high dose regimens.
Lung Cancer 13:137–143
Numerous studies show the effect on survival and Bezjak A, Dixon P, Brundage M et al (2002) Randomized
improvement in quality of life and disease-related phase III trial of single versus fractionated thoracic radiation
symptoms. Treatment with chemotherapy should be in the palliation of patients with lung cancer (NCIC CTG
restricted to patients with a reasonably good perfor- SC.15). Int J Radiat Oncol Biol Phys 54:719–728
Bradley JF, Ieumwananonthachai N, Purdy JA et al (2002)
mance status (WHO PS B 2). Most patients with Gross tumor volume, critical prognostic factor in patients
advanced non-small cell lung cancer will therefore be treated with three-dimensional conformal radiation therapy
offered chemotherapy as the first-line treatment. for non-small-cell lung carcinoma. Int J Radiat Oncol Biol
However, chemotherapy can generate toxicity and not Phys 52:49–57
Brundage MD, Davies D, Mackillop WJ (2002) Prognostic
all patients are considered fit. For these patients pri- factors in non-small cell lung cancer. Chest 122:1037–1057
mary palliative thoracic radiotherapy is a good option. Dehing-Oberije C, de Ruysscher D, van der Weide H et al
Palliative radiotherapy can also be offered to patients (2008) Tumor volume combined with number of positive
Palliative External Beam Thoracic Radiation Therapy of Non-Small Cell Lung Cancer 459

lymph node stations is a more important prognostic factor Council randomised trial of palliative radiotherapy with
than TNM stage for survival of non-small-cell lung cancer two fractions or ten fractions. Br J Cancer 63:265–270
patients treated with (chemo)radiotherapy. Int J Radiat Nestle U, Nieder C, Walter K et al (2000) A palliative
Oncol Biol Phys 70:1039–1044 accelerated irradiation regimen for advanced non-small-cell
Erridge SC, Gaze MN, Price A et al (2005) Symptom control lung cancer vs conventionally fractionated 60 Gy: results of
and quality of life in people with lung cancer: a randomised a randomized equivalence study 48:195–203
trial of two palliative radiotherapy fractionation schedules. NSCLC Meta-analysis collaborative group (2008) Chemother-
Clin Oncol 17:61–67 apy in addition to supportive care improves survival in
Fairchild A, Harris K, Barnes E et al (2008) Palliative thoracic advanced non-small-cell lung cancer: A systematic review
radiotherapy for lung cancer: a systematic review. J Clin and meta-analysis of individual patient data from 16
Oncol 26:4001–4011 randomized controlled trials. J Clin Oncol 26:4617–4625
Falk S, Girling DJ, White RJ et al (2002) Immediate versus Perez CA, Stanley K, Grundy G et al (1982) Impact of
delayed palliative thoracic radiotherapy in patients with irradiation technique and tumor extent in tumor control and
unresectable locally advanced non-small cell lung cancer survival of patients with unresectable non-oat cell carci-
and minimal thoracic symptoms: randomised controlled noma of the lung. Cancer 50:1091–1099
trial. BMJ 325:465–468 Rees GJG, Devrell CE, Barley VL et al (1997) Palliative
Hopwood P, Stephens RJ (1995) Symptoms at presentation for radiotherapy for lung cancer: two versus five fractions. Clin
treatment in patients with lung cancer: implications for the Oncol (R Coll Radiol) 9:90–95
evaluation of palliative treatment. Br J Cancer 71:663–666 Reinfuss M, Glinski B, Kowalska T et al (1999) Radiothérapie
Kirkpatrick JP, van der Kogel AJ, Schultheiss TE (2010) du cancer bronchique non á petites cellules de stade III
Radiation dose-volume effects in the spinal cord. Int J Rad inopérable asymptomatique. Résultats définitifs d’un essai
Oncol Biol Phys 76:suppl S42–S49 prospectif randomisé (240 patients). Cancer Radiother
Kramer G, Wanders SL, Noordijk EM et al (2005) Results of 3:475–479
the Dutch National Study of the palliative effect of Reinfuss M, Mucha-Malecka A, Walasek T et al (2011)
irradiation using two different treatment schemes for non- Palliative thoracic radiotherapy in non-small cell lung
small-cell lung cancer. J Clin Oncol 13:2962–2970 cancer. An analysis of 1,250 patients. Palliation of
Lester JF, Macbeth FR, Toy E, Coles B et al (2006) Palliative symptoms, tolerance and toxicity. Lung Cancer 71:344–
radiotherapy regimens for non-small cell lung cancer. 348
Cochrane Database Syst Rev 18(4):CD002143 Senkus-Konefka E, Dziadziuszko R, Bednaruk-Mlynski E et al
Lutz S, Norrell R, Bertucio C et al (2001) Symptom frequency (2005) A prospective randomised study to compare two
and severity in patients with metastatic or locally recurrent palliative radiotherapy schedules for non-small-cell cancer
lung cancer: a prospective study using Lung Cancer (NSCLC). Br J Cancer 92:1038–1045
Symptom Scale in a Community Hospital. J Palliat Med Simpson JR, Francis ME, Perez-Tamayo R et al (1985)
4:157–165 Palliative radiotherapy for inoperable carcinoma of the
Mac Manus MP, Matthews JP, Wada M et al (2005) lung: final report of a RTOG multi-institutional trial. Int J
Unexpected long-term survival after low-dose palliative Radiat Onc Biol Phys 11:751–758
radiotherapy for nonsmall cell lung cancer. Cancer Sundstrøm S, Bremnes R, Aasebø U et al (2004) Hypofrac-
116:1110–1116 tionated palliative radiotherapy (17 Gy per two fractions) in
MacBeth F, Wheldon TE, Girling DJ et al (1996) Radiation advanced non-small-cell lung carcinoma is comparable to
myelopathy: estimates of risk in 1,040 patients in three standard fractionation for symptom control and survival: a
randomized trials of palliative radiotherapy for non-small national phase III trial. J Clin Oncol 22:801–810
cell lung cancer. The Medical Research Council Lung Sundstrøm S, Bremnes R, Brunsvig P et al (2005) Immediate or
Cancer Working Party. Clin Oncol (R Coll Radiol) 8:176– delayed radiotherapy in advanced non-small cell lung
181 cancer (NSCLC)? Data from a prospective randomised
Maher EJ, Coia L, Duncan G et al (1992) Treatment strategies study. Radiother Oncol 75:141–148
in advanced and metastatic lung cancer: differences in Sundstrøm S, Bremnes R, Brunsvig P et al (2006) Palliative
attitude between the USA, Canada and Europe. Int J Radiat thoracic radiotherapy in locally advanced non-small cell
Oncol Biol Phys 23:239–244 lung cancer: can quality-of-life assessments help in selec-
Medical Research Council Lung Cancer Working Party (1992) tion of patients for short- or long-course radiotherapy? J
A Medical Research Council (MRC) randomised trial of Thorac Oncol 1:816–824
palliative radiotherapy with two fractions or a single Teo P, Tai TH, Choy D et al (1987) A randomized study on
fraction in patients with inoperable non-small cell lung palliative radiation therapy for inoperable non small cell
cancer (NSCLC) and poor performance status. Br J Cancer carcinoma of the lung. Int J Radiat Onc Biol Phys 14:867–
65:931–941 871
Medical Research Council Lung Cancer Working Party (1996) Yan S, Sun X Li MH et al (2007) A randomized study of
Randomised trial of palliative 2-fraction versus more involved field irradiation versus elective nodal irradiation in
intensive 13-fraction radiotherapy for patients with inoper- combination with concurrent chemotherapy for inoperable
able non-small cell lung cancer and good performance stage III non small cell lung cancer. Am J Clin Oncol
status. Clin Oncol 8:167–175 30:239–244
MRC Lung Cancer Working Party (1991) Inoperable non-
small-cell lung cancer (NSCLC): a Medical Research
Intraoperative Radiotherapy in Lung
Cancer: Methodology (Electrons
or Brachytherapy), Clinical Experiences
and Long-Term Institutional Results
Felipe A. Calvo, Javier Aristu, Sergey Usychkin, Leire Arbea,
Rosa Cañón, Ignacio Azinovic, and Rafael Martinez-Monge

Contents 8 Montpellier Regional Cancer Centre


Experience ................................................................ 466

1 Introduction.............................................................. 462 9 The Allegheny University Hospital


of Philadelphia Experience..................................... 467
2 Tissue Tolerance Studies of IORT ........................ 462
10 Madrid Institute of Oncology (Grupo IMO)
3 Technical Considerations: IOERT ........................ 463 Experience ................................................................ 467
4 Clinical Indications: IOERT .................................. 464 11 USP Hospital San Jaime Experience .................... 467
5 International Intraoperative Electron 12 The University Clinic of Navarra Experience ..... 468
Radiotherapy (IOERT) Clinical Experiences
and Results ............................................................... 464 13 International LDR-IORT and
HDR-IORT Clinical Experiences and Results ..... 470
6 National Cancer Institute Experience................... 464
14 Stage I–II Disease .................................................... 471
7 University Medical School of Graz Experience... 465
15 Stage III Disease ...................................................... 472
16 Superior Sulcus Tumors (SST) .............................. 472
17 Summary and Final Considerations...................... 473
References.......................................................................... 475

F. A. Calvo (&)
Abstract
Department of Oncology,
University Hospital Gregorio Marañón, Intraoperative radiotherapy is a feasible technical
University Complutense, Madrid, Spain modality to improve precision and dose-escalation
e-mail: fcalvo.hgugm@salud.madrid.org in high-local risk lung cancer patients. Methodology
J. Aristu  L. Arbea is described regarding the use of high-energy
Department of Oncology, University Clinic, electron beams or brachytherapy. Results of normal
University of Navarre, Pamplona, Spain
tissue tolerance in experimental animal models and
S. Usychkin in clinical experiences are analyzed in detail.
Department of Radiotherapy,
Instituto Madrileño de Oncología,
Characteristics of clinical experiences using IORT
Madrid, Spain electrons or brachytherapy are reported and clinical
R. Cañón  I. Azinovic
outcome results are discussed. Ten IORT brachy-
Department of Radiation Oncology, therapy and six electron-based publications are
Hospital San Jaime, Torrevieja, Spain identified proving the adaptability of IORT to the
R. Martinez-Monge clinical-therapeutic scenario of lung cancer, its
Department of Radiation Oncology, feasibility and the promotion of high local control
Clinica Universitaria de Navarra, rates in the context of dose-escalation trials.
Pamplona, Spain

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 461
DOI: 10.1007/174_2011_286, Ó Springer-Verlag Berlin Heidelberg 2011
462 F. A. Calvo et al.

IORT has been integrated into multi-disciplinary


1 Introduction programs as a boosting modality that completes the
total dose given with fractionated external beam
Lung cancer is the leading cause of cancer-related radiation therapy (ISIORT 1998). This treatment has
mortality worldwide, with nearly 1.4 million deaths the advantage of the radiobiological effects of frac-
each year (Jemal et al. 2010). Lung cancer is diag- tionation over the primary volume that includes the
nosed at an advanced stage in a majority of patients, primary tumor and the draining areas while the tumor
which is the primary reason behind the high mortality bed is boosted with single-dose electrons.
rate associated with this disease (Ramalingam et al. The current review describes methodology and
2011). At present 5-year overall survival in operable clinical results of retrospective analyses including the
or marginally operable locally advanced disease is prognostic factors related with local control and sur-
about 20–25% (Hansen and Roach 2010). vival in large institutional experiences generated in
There is a well-known dose response relationship NSCLC patients treated with IOERT component
in non-small-cell lung cancer (NSCLC). In a study of within a multi-disciplinary treatment program.
Rengan et al. (2004) the median survival time for
patients treated with 64 Gy or higher was 20 months
versus 15 months for those treated with less than 2 Tissue Tolerance Studies of IORT
64 Gy and a 10 Gy increase in dose resulted in a
36.4% decreased risk of local failure. A phase I–II The tolerance of mediastinal structures to IORT with
RTOG 9311 study confirmed a safety of dose-esca- high-energy electron beams (IOERT)ental animal
lation to 83.8 Gy with three-dimensional conformal studies. In a dose-escalation study (Barnes et al. 1987)
techniques (Bradley et al. 2005). In a recently pub- delivering 20, 30 and 40 Gy to two separated intra-
lished study (Dong-Soo et al. 2011) showed that thoracic IOERT fields which included collapsed right
clinical tumor response after concurrent chemoradia- upper lobe, esophagus, trachea, phrenic nerve, right
tion in locally advanced, recurrent and postoperative atrium and blood vessels, pathologic changes were
gross residual NSCLC was the only significant observed at 30 Gy in the trachea and esophagus, with
prognostic factor for overall survival. severe ulceration and peribronchial and perivascular
Pattern of failure data show that 40–70% of chronic inflammation in the normal lung. At dose of
the patients with non-small-cell lung cancer stages 20 Gy medial and adventitial fibrosis, obliterative
II–IIIB are expected to relapse locally (Kumar et al. endarteritis of the vasa vasorum, and severe coagu-
1996). Recently dose-dependent pattern of failure in a lative necrosis were observed. Acute pneumonitis was
NSCLC was demonstrated. In a study of Sura et al. seen at all doses, and changes in the contralateral lung
(2008) 75% of patients with NSCLC who received were detected using 12 MeV electrons.
\60 Gy had failure within GTV and 25% had disease De Boer et al. (1989) studied the effects of 20, 25
relapse at the GTV margin while among patients who and 30 Gy in mediastinal structures. The bronchial
received C60 Gy 33 and 61% had relapse within stump healed in all dogs. Severe tissue damage was
GTV and at the margin of GTV, respectively. seen at all doses and included bronchovascular and
Local control in NSCLC continues being an esophagoaortic fistulas and esophageal stenosis.
unresolved issue and the introduction of new radiation At the National Cancer Institute, an experimental
techniques to intensify the local dose is justified. program evaluated the tolerance of surgically
Intraoperative radiation (IORT) is a sophisticated manipulated mediastinal structures to IOERT in 49
radiation modality well explored in the treatment adult foxhounds. Tolerance of normal tissues was also
of abdominopelvic tumors but is scarcely used in evaluated in a limited phase I clinical trial (four
thoracic tumors. The therapeutic gain in IORT pro- patients with stage II or III NSCLC). Normal healing
cedures is obtained with the displacement of radio- of the bronchial stump was found after pneumonec-
sensitive organs away from the electron beam or with tomy at IOERT doses of 20, 30 and 40 Gy, but there
the shielding of fixed structures with lead sheets. were late changes with tracheobronchial irradiation
Target definition is done after the surgical resection damage at all doses (5–10 months after treatment).
jointly with the thoracic surgery team. Two out of four patients receiving 20 Gy developed
Intraoperative Radiotherapy in Lung Cancer 463

Table 1 Clinical and pathologic findings observed in animal experimental models (Barnes et al. 1987; De Boer et al. 1989;
Tochner et al. 1992; Sindelar et al. 1992; Zhou et al. 1992; Kritskaia et al. 2006)
IORT doses Bronchial stump Esophageal damage Lung damage Pathologic changes in
heart and vessels
20 Gy Normal healing Transient mild dysphagia Mild Moderate
30 Gy Normal healing Chronic ulcerative esophagitis Moderate Moderate-severe
40 Gy Normal healing Esophageal perforation Severe Severe
Esophageal stricture

esophageal ulceration at 6 months without late stric- cisplatin (Kritskaia et al. 2006). No degenerative
ture. In dogs given 30 and 40 Gy, esophageal damage changes in the bronchial epithelium were found
was severe (esophagoaortic fistula and stenosis) and 2 weeks after IORT. Basal cell proliferation was
one dog developed carinal necrosis. The same insti- observed, goblet cells were reduced in size and the
tution reported the results of five dogs reserved for basement membrane was thickened and twisted.
long-term studies and one stage II NSCLC patient Epithelial reparation due to pronounced local basal
alive at 5 years. They conclude that IOERT in the cell proliferation was observed 3 months later. A year
mediastinum may be safe at dose levels that do not later, the mucosa was covered with the multi-nuclear
exceed 20 Gy (Tochner et al. 1992). cylindrical epithelium and the cover of ciliated cells
Additional experimental analysis of canine esoph- was preserved. The functional activity of goblet cells
agus tolerance to IOERT has been reported by the was in the normal range and scanty lympho-plasmo-
NCI investigators (Sindelar et al. 1992). After right cytic infiltration was found in the stroma. In patients
thoracotomy with mobilization of the intrathoracic treated with IORT without radiosensitization, the
esophagus, IOERT was delivered to include a 6 cm damaged epithelium was regenerated due to the
esophageal segment using a 9 MeV electron beam with reserved cells coming from the damaged margins with
escalating single doses of 0, 20 and 30 Gy. Dogs were the formation of an epidermoid regenerative layer and
followed clinically with endoscopic and radiologic subsequent cell differentiation. Moderate sclerosis
studies and were electively sacrificed at 6 weeks or 3, occurred in the stroma. A year later the bronchial
12 or 60 months after treatment. Transient mild dys- epithelium was characterized by moderate goblet cell
phagia and mild esophagitis was observed in all dogs hyperplasia with preserved functional activity. The
receiving 20 Gy, without major clinical or pathological authors concluded that IORT caused mucosal damage
sequelae except in one dog that developed achalasia as alteration, dystrophy and desquamation of the
requiring a liquid diet. At a dose of 30 Gy, changes in epithelium. Subsequently, the bronchial epithelium
the esophagus were pronounced with ulcerative recovered through reparative regeneration.
esophagitis and chronic ulcerative esophagitis inducing Based on these data, active clinical programs using
gross stenosis after 9 months. thoracic IOERT agree that 20 Gy is the upper single-
Zhou et al. (1992) analyzed the acute responses of dose limit that can be safely tolerated by mediastinal
the mediastinal and thoracic viscera in nine canines that and thoracic viscera (Table 1) with IOERT alone.
were sacrificed after they received single IOERT doses There are no reported experimental normal-tissue
of 25, 35 and 45 Gy. No pathological changes were tolerance studies of IOERT used in combination with
found in the spinal cord and vertebra. Microscopic EBRT.
examination of trachea, esophagus and lung showed
mild or severe histological changes at 30 days at the
level of 25 Gy versus 35–45 Gy, respectively. Severe 3 Technical Considerations: IOERT
and unrepaired histologic changes were found in the
heart and aorta receiving 35–45 Gy. IOERT requires the adaptation of linear accelerator
Morpho-functional changes in the bronchial with multi-energetic electron beam capability (ener-
mucosa were studied in 33 patients with stage III gies recommended from 6 to 20 MeV), through the
NSCL treated with 15 Gy IORT with or without development of specially designed applicators for
464 F. A. Calvo et al.

4 Clinical Indications: IOERT

IOERT at the time of thoracotomy for a surgical


approach to lung cancer has been employed in three
different situations:
– Treatment of unresectable hiliar and/or mediastinal
disease
– Treatment of postresected residual disease (chest
wall, mediastinum and/or bronchial stump)
– Adjuvant treatment of mediastinum.
Conceptual indications for IOERT in thoracic
surgery have been the treatment of residual disease at
Fig. 1 Equipment used in a IOERT procedure: gantry adapter
the primary site and/or nodal regions, or adjuvant
with mirror-carrier (A); intermediate element (B) transparent treatment of high risk of recurrence without proven
methacrylate applicator with a metric reference (C); distal cancer residue after induction therapy and surgery.
section of a beveled applicator (D) IOERT is a super-selective radiation boost component
available for integration in conventional radiotherapy
electron-beam conformation (cone sizes recommended programs for lung cancer. Lung parenchyma is the
from 5 to 12 cm diameter) (Fig. 1). The clinical pro- normal tissue that may benefit the most from IOERT.
gram combines the efforts of surgeons, anesthesiolo- Esophagus, trachea, aorta and heart are difficult to
gists, physicists and radiation oncologists to adequately displace from the IOERT beam, particularly in the
select patients for IOERT indications, perform the treatment of mediastinal regions or left lower chest
surgical procedure (tumor resection plus normal-tissue cavity. In the case that the bronchial stump is included
protection), transport and monitor the patient during in the IOERT field, tissue coverage with a vascular-
intraoperative irradiation and finally decided the radio- ized pleural or pericardial flap is recommended to
therapeutic parameters for treatment prescription promote bronchial healing.
(Figs. 2, 3). In general, IOERT during lung cancer
surgery involves the coordination of 10–15 health
professionals, prolongs the surgical time approxi- 5 International Intraoperative
mately 30–45 min (depending upon transportation Electron Radiotherapy (IOERT)
time), and induces a 2 h gap of time availability in the Clinical Experiences and Results
linear accelerator for outpatient treatment.
Miniaturized and mobile linear accelerator such as The clinical experience of IOERT in lung cancer is
the Mobetron operates at energies of: 4–12 MeV, still limited and the available data regarding treatment
using two different dose rates: 2.5 and 10 Gy/min. of NSCLC were obtained in phase I–II trials in a
These energies are sufficient to achieve a 4 cm depth small series of patients. Abe and colleagues in the
of relative uniform beam penetration with acceptable initial Japanese experience did not use IOERT in lung
dose homogeneity (±5%) (Merrick and Dobelbower neoplasms because of the early systemic dissemina-
2003) (Fig. 4). tion of disease (Abe and Takahashi 1981).
The unit is mounted on a gantry and cantilevered
with a beam after it passed through the patient. This is
the principal feature that allows this unit to be used in 6 National Cancer Institute
any operating room without additional shielding. Experience
In addition to C-arm rotation the unit can rotate in and
out and in orthogonal planes. This design increases its Based on a previous canine experimental model
versatility in setting up patients for treatment and involving the use of pneumonectomy and IOERT
reduces the amount of time needed to align the radiation doses of 0, 20, 30 and 40 Gy, a limited phase I
field with the applicator (Biggs et al. 2003) (Fig. 5). National Cancer Institute (NCI) clinical trial
Intraoperative Radiotherapy in Lung Cancer 465

Fig. 2 Different general


views of thoracic IOERT with
electrons in superior sulcus
tumor (1, 2), upper
mediastinum tumor (3) and
left hilum tumor (4).

demonstrated considerable toxicity with 25 Gy of with N0 disease. The radiosensitive mediastinal


IOERT to two separated fields encompassing the structures such as the heart, spinal cord, esophagus and
superior and inferior mediastinum following pneu- large vessels could be mobilized or protected from the
monectomy (Pass et al. 1987). Early complications IOERT beam by shielding maneuvers.
were described in three out of four patients: one case The response rates in 14 evaluable patients
of bronchial stump dehiscence, one broncho-pleural 18 weeks after they completed IOERT and EBRT were
fistula and one case of reversible esophagitis. Three excellent with three complete responses (21%) and 10
patients with late complications showed one case of partial responses (71%). Ten patients are alive and well
irreversible radiation esophagitis. Only one long-term at a range of 5–20 months (median 12 months).
survivor is free from disease (more than 3 years). The The same institution updated the results of this
retrospective analysis of toxic events detected over- program in two consecutive studies (Arian-Schad
lapping of the fields in one toxic case. This study et al. 1990; Smolle Juettner et al. 1994). The IOERT
recognized the feasibility of IOERT during lung procedure was generally well tolerated, but fatal intra-
cancer surgery and recommended a decrease in the bronchial hemorrhage related to IOERT occurred in
IOERT dose to 15–20 Gy. two cases with tumor involvement of the pulmonary
artery. Local failure was seen in three patients and the
5-year overall and recurrence-free survival rates were
7 University Medical School of Graz 15 and 53%, respectively.
Experience An expanded series from the University of Graz has
been recently published (Jakse et al. 2007). Fifty-two
Combined IOERT (10–20 Gy) and postoperative patients with predominantly pathological stage I
EBRT (46–56 Gy) were used in 21 inoperable tumors NSCLC (76%) with limited pulmonary reserve (med-
at the University Medical School of Graz (Austria) ian FEV1: 1,3) were treated with surgery, IORT
(Jeuttner et al. 1990). The analysis included 12 patients (median dose 20 Gy) and EBRT (median dose 46 Gy).
466 F. A. Calvo et al.

Fig. 3 Simulation for applicator selection (size, beveled angle, a Pancoast’s tumor. The target volume includes the tumor bed
positioning and maneuvers for normal-tissue protection) after region (posterior and superior chest wall and paravertebral space),
right superior lobectomy. The IOERT target volume includes right and the remaining normal lung is mobilized out of the intraop-
mediastinum and bronchial stump; the remaining normal lung is erative field (2). IOERT applicator positioning during exploratory
mobilized out of the electron field (1). Postresection simulation for thoracotomy for an unresectable right-lobe NSCLC (3)

survival than males. Causes of death were unrelated to


tumor in 17% and tumor related in 54% patients.
Two patients died from second cancers and 25% are
alive without evidence of tumor progression. Overall
loco-regional tumor control was 73% at 12 months and
68% at 24 and 36 months, respectively. IORT and
EBRT were well tolerated without serious treatment
related acute or late side effects.

8 Montpellier Regional Cancer


Centre Experience

Fig. 4 Mobetron mobile linear accelerator in the operating room The Centre Regional De Lutte Contre Le Cancer In
Montpellier (France) reported results in 17 patients:
The actuarial overall survival and disease-specific three stage I, seven stage II and seven stage IIIA (Carter
survival at 3 years were 37 and 48%, respectively. et al. 2003). The treatment protocol involved the use of
Females had a significantly better disease-specific IOERT with doses in the range of 10–20 Gy and 45 Gy
Intraoperative Radiotherapy in Lung Cancer 467

Fig. 5 Mobetron set-up for intraoperative irradiation

EBRT in 20–25 fractions with or without a 3 week rest for the alive patients is 33 months. Patterns of relapse
period following a complete surgical excision. Micro- have shown 3 (14%) thoracic and 12 (55%) systemic.
scopic residual disease in the mediastinal nodes or Actuarial 5-year survival was 33%.
pleura-chest wall was seen in 12 and 5 patients,
respectively. The median follow-up time for the entire
group of patients alive was 59 months, with follow-up 10 Madrid Institute of Oncology
ranging from 40 to 120 months. (Grupo IMO) Experience
Disease control and survival results were as follow.
Local control was obtained in 13 out of 17 patients In the Instituto Madrileño de Oncología (Grupo IMO)
(76%) and central recurrence in the IOERT field has in Madrid from February 1992 to July 1997, 18 patients
been demonstrated in four patients. Three patients are with stage III non-small-cell lung cancer (11 Pancoast’s
alive without disease at 5.5, 8 and 11 years. Fourteen tumors) received IOERT as a part of a multi-disci-
patients are dead: seven from distant metastases, four plinary program including surgical resection in all
from loco-regional recurrence, one patient developed cases, chemotherapy in 13, preoperative EBRT in 7,
a second cancer, and two patients had a local recur- and postoperative EBRT in 7. Tumor residue at the time
rence in the EBRT field. The median survival time for of surgery was macroscopic (gross) in eight cases. The
the entire group was 36 months and the actuarial median survival time for the entire series is 14 months.
survival rate is 18% projected at 11 years. Intra-thoracic recurrence has been identified in two
patients. Five-year actuarial survival is projected
as 22% (cause-specific 33%). Long-term toxicity
9 The Allegheny University Hospital observed included neuropathy (two cases) and esoph-
of Philadelphia Experience ageal structure (one case) (Calvo et al. 1999).

This unique experience in the United States was pre-


liminarily reported in 1994 (Fisher et al. 1994). The last 11 USP Hospital San Jaime Experience
update (Aristu et al. 1999) includes 21 patients treated
from 6/92 to 9/97 as a part of a pilot feasibility expe- Between June 2004 and October 2008 in the USP
rience for stage I (n = 1), II (n = 2) and III (n = 18) Hospital San Jaime in Torrevieja, Spain, eight patients
NSCLC patients managed by surgical resection, have been treated with IORT using the Mobetron
IOERT (10 Gy), and EBRT (45.0–59.4 Gy, 16 preop- mobile linear accelerator: two woman and six men,
eratively and 5 postoperatively). Chemotherapy was with a median age of 53 years (range 45–66 years). All
administered to all patients. The median survival time patients had locally-advanced non-small-cell lung
468 F. A. Calvo et al.

cancer (NSCLCs): stage IIA (n = 1), stage IIIA IOERT as a treatment component of multidisciplinary
(n = 1), stage IIIB (n = 4), stage IV (n = 1, a woman management at the University Clinic of Navarra in
with T2N2 and brain metastases) and one patient with Pamplona (Spain) (Calvo et al. 1990, 1991, 1992;
local relapse after surgery and radiotherapy. Aristu et al. 1997, 1999; Martínez-Monge et al. 1994).
All patients received preoperative radiochemo- Twenty-two patients were treated with surgery,
therapy: 50 Gy with conventional fractionation, IOERT and postoperative EBRT, 82 patients received
except two patients: one patient with local relapse neoadjuvant chemotherapy and were treated depend-
after radiochemotherapy and then treated with hypo- ing on tumor response as follows: 46 responders with
fractionation (10 Gy for four sessions, 2 days a week) respectable tumor were managed with surgery, IO-
and one patient stage IV, who received 66 Gy with a ERT and postoperative EBRT, non-responders with
radical intention and systemic chemotherapy. There unresectable disease (17 patients) or Pancoast’s tumor
were three ypT0, five ypTmicroscopic, five ypN0, one (19 patients) received preoperative chemoradiother-
ypNmicro and two ypN1 pathologic responses in the apy, surgery and IOERT boost.
surgical specimens. The neoadjuvant chemotherapy consisted of cis-
The median dose of IORT administered was 10 Gy platin 120 mg/m2 i.v. on day one, mitomycin C 8 mg/
(range 6–10 Gy), using a cone of 4.5 cm of diameter m2 i.v. day one and vindesine 3 mg/m2 (maximum
(range 3.5, 6 cm), 9 meV (range 4–9 MeV) electron dose 5 mg/m2) i.v. on days one and fourteen (MVP)
beam and with a median time for the procedure of or the same treatment regimen where the cisplatin
35 min, range: 15–60 min. administration was replaced by intraarterial carbo-
At a median follow-up of 36.5 months (range platin 150 mg/m2. The cycles of chemotherapy were
2–66 months): one patient is lost for follow-up, two repeated every 28 days for 3–5 treatments until
died free of disease, one died with local and brain achieved maximum response (3–5 cycles). Patients
relapse, three are alive free of disease 36, 37 and who documented a clinical response or with stable
53 months after the IORT, and one is alive with disease and considered resectable were referred to
pleural and mediastinal relapse 2 years after the IORT surgical resection including the primary tumor and
and 1 year after suprarenalectomy for metastasis. mediastinal lymphadenectomy 4–5 weeks after the
Two patients died 1.5 months after the IORT were: last cycle of neoadjuvant chemotherapy. The bron-
the first patient who had received 66 Gy of radio- chial stump was protected with a pleural or pericardial
therapy died for lung complication, and the second flap in order to prevent anastomotic leak. After sur-
patient with local relapse after radiochemotherapy gical resection IOERT was applied over the surgical
and treated with hypofractionation died from massive bed, hiliar and mediastinal regions depending on the
bleeding. tumor location. Total dose administered varied
The median overall survival is 22 months and the between 10 and 15 Gy depending on the amount of
disease-free survival is 12 months. Patterns of failure residual tumor. A detailed description of the IOERT
are: one patient with local relapse was rescued with methodology for thoracic tumors has been published
reirradiation. Six months after he progressed with previously (Calvo et al. 1990, 1992; Aristu et al.
brain metastases and died with disease. The second 1997; Martínez-Monge et al. 1994). Postoperative
patient had adrenal gland metastases 11 months after external beam radiation therapy was started four to
surgery which were rescued with suprarenalectomy 5 weeks after surgical resection. Treatment was
and 13 months after he developed pleural and medi- delivered with a linear accelerator employing AP–PA
astinal relapse. technique to encompass the treatment volume which
included bronchial stump, ipsilateral hilum, the
bilateral mediastinal and supraclavicular lymph
12 The University Clinic of Navarra nodes. A dose of 46 Gy in 23 fractions was applied.
Experience Tumors that were not considered resectable after
neoadjuvant chemotherapy were treated with preop-
During the period from November 1984 to November erative external beam radiation therapy using the
1993 104 patients with histologically confirmed non- same total dose and fractionation than with postop-
small-cell lung cancers stage III were treated with erative external beam radiation therapy described
Intraoperative Radiotherapy in Lung Cancer 469

Table 2 Patterns of failure Surgical residue Local controla Distant failureb


according to disease stage and
surgical residue in the Micro/Absent
University Clinic of Navarra IIIA 18/24 (75%) 7/24 (29)
Experience
IIIB 4/14 (29%) 4/14 (29)
Pancoast’s tumors 11/12 (92%) 2/12 (17)
Macroscopic/Unresected
IIIA 3/7 (43%) 6/7 (86)
IIIB 7/30 (23%) 12/30 (40)
Pancoast’s tumors 5/5 (100%) 1/5 (20)
a
No local or distant failure
b
Distant failure alone or distant and local failure

above. All patients received concurrent chemotherapy fistulas and two pulmonary hemorrhages. The first
with preoperative radiation using the same chemother- broncho-pleural fistula occurred in a lobectomized
apy combination used as neoadjuvant or with cisplati- patient, in whom the bronchial stump was not included
num 20 mg/m2 or carboplatinum 55 mg/m2 combined into the IOERT field. Another patient died 3 months
with five fluorouracil 1,000 mg/m2 (maximum daily after surgery due to a broncho-pleural fistula in a
dose 1,500 mg) during three to five days over the first microscopically tumor involved bronchial stump.
and last week of external beam radiation therapy. Four One patient developed fatal massive hemoptysis at
to six weeks after the completion of the preoperative 2 months following IOERT because of pulmonary
chemoradiation course the patients were referred for artery rupture. This latter patient had prior hemoptysis
surgical resection and IOERT, when feasible. and a left hiliar unresected tumor treated by tumor
The local control rates observed in patients with exposure and 15 Gy (20 MeV) IOERT plus 46 Gy
\microscopic residual disease (R0 or R1 resection) postoperative EBRT. The autopsy study showed a
were 18/24 (75%), 4/14 (29%) and 11/12 (92%) for necrotic cavity in the primary tumor with no viable
stage IIIA, IIIB and Pancoast’s tumors, respectively. residual tumor cells and a fistulous tract communicat-
Local control in patients with macroscopic residual ing between the pulmonary artery and the bronchial
disease (R2 resection) were 3/7 (43%), 7/30 (23%) tree. A non-resected patient treated with three cycles of
and 5/5 (100%) for stage IIIA, IIIB and Pancoast’s MVP regimen, preoperative EBRT (44 Gy) and IORT
tumors, respectively. Table 2 describes the patterns of of 15 Gy died early in the postoperative period from
failure according to the treatment group. pulmonary hemorrhage.
At the time of analysis 16 patients (15%) were alive Esophagitis grade 3–4 was noted in 26 (25%)
and free of disease. Five-year OS for the entire group patients and esophageal damage with ulcerated or
was 40% for stage IIIA and 18% for stage IIIB patients necrotic tissue was observed in two patients. One out of
(P = 0.01). Five-year disease free survival (DFS) two patients who developed esophageal ulcer died
regarding amount of residual disease is as follows: 69% 8 months after surgery from fatal hemorrhage. This
and 42% for microscopic (R1) or no residual disease patient had a T4 tumor infiltrating the descending
(R0) for stages IIIA and IIIB, respectively and 58 and portion of the aorta and the esophagus. He was treated
41% for macroscopic (gross) residual disease (R2) for with three cycles of MVP chemotherapy regimen,
stages IIIA and IIIB, respectively. Anecdotally, 19 preoperative EBRT (46 Gy), surgery (atypical resec-
patients survived more than 5 years after IOERT with a tion plus chest wall resection), and 10 Gy IORT boost
follow-up range from 64 to 107 months. Among (12 MeV). No viable microscopic tumor was encoun-
patients surviving more than 5 years there were tered in the resected specimen and the necropsy find-
3 second tumors (colon, esophagus and head and neck) ings revealed a connection between the esophagus and
and one cancer-unrelated death. the aorta without histological evidence of tumor cells.
Regarding treatment toxicity and complications, Symptomatic radiation acute pneumonitis was
four patients died in the postoperative period due to observed in six patients. Seven patients were diag-
possible IOERT-related toxicity: two broncho-pleural nosed with severe long-term fibrosis and required
470 F. A. Calvo et al.

Table 3 IOERT international clinical experiences in NSCLC


Authors (reference) Number Stage Treatment protocol Local 5-year
of control survival
patients
University Medical School of Graza 24 12 I IORT 10–20 Gy 19/23 15%
(Smolle Juettner et al. 1994) 1 II + (83%)
10 EBRT 46–56 Gy
IIIA
Montpellier Regional Cancer Centre 17 3I S ? IORT (10–20 Gy) 13/17 18%
(Carter et al. 2003) 7 II + (76%)
7 IIIA EBRT 45 Gy
University Clinic of Navarra in 104 19 Multidisciplinary treatment (see text) 48/92 40% (IIIA)
Pamplona IIIA with IORT 10–20 Gy ? EBRT (52%) 18% (IIIB)
(Calvo et al. 1990, 1991, 1992; Aristu (N0) (46 Gy) + CT
et al. 1997, 1999; Martínez-Monge 29
et al. 1994) IIIA
(N2)
56
IIIB
The Allegheny University Hospital of 21 1I Neoadjuvant CT ± preop EBRT 18/21 33%
Philadelphia 2 II + (86%)
(Aristu JJ, Fisher S, Calvo FA et al. 15 S ? IORT ± postop EBRT
1999) IIIA
3 IIIB
Madrid Institute of Oncology (Grupo 18 11 Neoadjuvant CT ± preop EBRT 16/18 22%
IMO) (Calvo et al. 1999) IIIA + (90%)
6 IIIB S ? IORT ± postop EBRT
1IV
USP Hospital San Jaime in Torrevieja 8 1 IIA Preoperative EBRT 50 Gy 6/8 Median OS
(Cañon R et al. 2008) 1 IIIA + (75%) 22 m, DFS
4 IIIB S ? IORT 6–10 Gy 12 m
1 IV
1 local
relapse
CT Chemotherapy
a
Inoperable patients

chronic cortico-therapy administration. Neurologic


toxicity was noted only in patients treated with 13 International LDR-IORT and
IOERT which included the thoracic apex or chest HDR-IORT Clinical Experiences
wall. Six patients developed transient neuropathy and Results
(four Pancoast’s tumors) with pain and paresthesia in
the superior ipsilateral extremity or chest wall. Severe Intraoperative brachytherapy using low-dose rate
infectious complications were seen in 11 patients. Six (LDR-IORT) or high-dose rate (HDR-IORT) is a
of these patients were diagnosed with simultaneous radiation treatment alternative in lung cancer patients
thoracic tumor progression coexisting with an who are technically operable but cannot tolerate the
abscess. operative procedure and the expected reduction in
A summary of NSCLC IOERT clinical experi- lung function after resection or conventional EBRT.
ences from different institutions is presented in LDR-IORT/HDR-IORT can be also used as a radia-
Table 3. tion boost technique in patients with residual disease
Intraoperative Radiotherapy in Lung Cancer 471

after chemoradiation or in previously irradiated could be demonstrated among patients with squamous
patients diagnosed with recurrent disease. versus adenocarcinoma, T1 versus T2 tumors or those
The LDR-IORT/HDR-IORT technique to be used who did or did not receive postoperative EBRT.
depends on tumor location and the volume of residual Fleischman et al. (1992) have published the results
disease after resection (R0, R1 and R2). Resectable of 14 medically inoperable stage I patients treated
but inoperable tumors, R2 resections and recurrent with I-125 implantation at thoracotomy. Doses ranged
tumors may be treated by a permanent implant using from 80 Gy at the periphery to 200 Gy at the
Iodine-125 (I-125) or Palladium-103 (Pd-103) seeds. center. There was one operative mortality and two
Unresectable chest wall lesions and R1 resections postoperative complications. With a minimum fol-
may be treated intraoperatively by either a temporary low-up of 1 year, the local control was 71% and the
Iridium-192 (Ir-192) implant or a permanent I-125 median survival was 15 months.
implant imbedded in absorbable polyglactin (vicryl) A retrospective multi-center study of 291 patients
sutures and directly sutured onto the target area with T1N0 disease was done comparing the outcomes
(d’Amato et al. 1998) or it may be treated by employing after sub-lobar resection (124 patients) and lobar
I-125 seeds imbedded into an absorbable gelatine resection (167 patients) (Fernando et al. 2005).
sponge (Gelfoam) plaque (Nori et al. 1995a). Periop- Brachytherapy (100 Gy to 120 Gy to a 0.5 cm depth)
erative high-dose-rate brachytherapy (PHDRB) using was used in 60 patients with sub-lobar resection. With
Ir-192 administered over the immediate postoperative a mean follow-up of 34.5 months, brachytherapy
period has been mainly used in R0–R1 tumor resec- decreased the local recurrence rate significantly
tions. Intraoperative implantation of plastic catheters among patients undergoing sub-lobar resection from
into the tumor bed after surgical resection for PHDRB 17.2 to 3.3%. There was no difference in survival
has several theoretical advantages over other types of between sub-lobar resection and lobar resection in
radiation boosting techniques, including: (i) accurate tumors smaller than 2 cm. However, for tumor rang-
real-time definition of the clinical target volume (CTV) ing from 2 to 3 cm, median survival was significantly
surrounding the tumor bed and other high risk areas better in the lobar resection group.
(with the assistance of the surgical team); (ii) CT scan- The experience of the New England Medical
based treatment planning; (iii) risk-adapted brachy- Center in Boston is based on the implantation of
therapy dose selection based upon the amount of radioactive I-125 seeds along the resection margin in
residual disease described in the final pathology report; 35 patients with stage I lung cancer treated with
and (iv) early delivery of fractionated radiation during limited resection (not candidates for lobectomy) (Lee
the immediate postoperative period. et al. 2003). Two patients developed local recurrence
at the resection margin and six patients developed
regional recurrences in the mediastinun or chest wall.
14 Stage I–II Disease The 5 year OS was 67 and 39% for patients with
T1N0 and T2N0 tumors, respectively.
The largest experience with IOBT has been published Investigators of the University of Pittsburgh
in patients with stage I–-II lung cancer who are unfit Cancer Institute reported a trial exploring the feasi-
for surgery and radical EBRT. The majority of the bility and outcomes of 125-I Vicryl mesh brachy-
studies are retrospective and come from single insti- therapy after sub-lobar resection (open or video-
tutions. The MSKCC experience has been reported by assisted thoracoscopic procedure) in stage I non-
(Hilaris and Mastoras 1998). The study included 55 small-cell lung cancer patients with poor pulmonary
patients treated with thoracotomy, intersticial I-125 function (Chen et al. 1999; Voynov et al. 2005). The
implantation ± moderate doses of EBRT. There were implant was introduced through the surgical incision
no operative or postoperative deaths. Locoregional and sutured to the visceral pleura. A prescribed dose
control at 5 years was 100% in T1N0 lesions, 70% of of 100–120 Gy was delivered to a volume within
patients with T2N0 tumors and 71% in T1-2N1 0.5 cm from the plane of the implant. There were four
tumors. The 5 year OS was 32% and DFS was 63%. local recurrences in the 110 patients treated and the
The median survival was better in patients with estimated 5 year local control, locoregional control,
cancer in the right lung but no difference in survival and OS rates were 90, 61, and 18%, respectively.
472 F. A. Calvo et al.

The same institution presented a later experience


15 Stage III Disease including 225 patients with thoracotomy and IOBT
when need in primary non-small-cell lung invading
In the University of Navarra investigators initiated a only the mediastinum (T3-4N0-2) (Burt et al. 1987).
prospective, nonrandomized, controlled Phase II The authors encountered a positive correlation
clinical trial to determine whether perioperative between prolongation of survival and extent of
high-dose-rate brachytherapy (PHDRB) using Ir-192 resection/IORT. Forty-nine patients had complete
administered over the immediate postoperative period resection without IORT and fared no better than a
is feasible and tolerable and may improve locore- cohort group of 33 patients underwent pulmonary
gional control rates in lung cancer patients with resection with simultaneous iodine-125 interstitial
residual disease after chemoradiation or recurrent implantation or iridium-192 delayed afterloading to
disease after previous radiation therapy (Valero et al. areas of unresectable primary or nodal disease. The
2007). In R0/R1 lung cancer resections the tumor bed median survival, 3 and 5 year survival was
was implanted with plastic catheters for PHDRB. The 17 months, 21 and 5%, respectively, with incomplete
brachytherapy dose was 4 Gy b.i.d. x 4–10 fractions resection; and 12 months, 22 and 22% with incom-
(16–40 Gy total dose). Selected technically unfeasible plete resection and brachytherapy. One hundred and
cases for PHDRB were treated using a silicone mold one patients underwent interstitial implantation
in which plastic catheters are inserted and a single without resection, with a median survival of
dose of 10–12.5 Gy was administered. Macroscopic 11 months, 3 year survival of 9%, and no 5 year
residual unresectable tumors (R2 resections) were survivors. The perioperative mortality was 2.7% and
implanted with I-125 or Pd-103 seeds to deliver a the nonfatal complication rate 13%.
minimum tumor dose of 90–110 Gy. Between 2001 Researchers in the New York Hospital Medical
and 2006 period, 20 patients have been treated, 15 Center of Qeens in New York investigated the safety,
patients had residual disease and 5 patients had recur- reproducibility and effectiveness of intraoperative
rent disease. Two patients developed grade three I-125 or Pd-103 Gelfoam plaque implant technique in
complication with thoracic abscess. Nine patients are 12 patients as a treatment complement for resected
alive, seven without disease, one without disease after stage III patients with positive surgical margin. All
radiosurgery for brain metastases and one patient is patients received preoperative or postoperative EBRT
alive with disease. The local, locoregional and systemic (45–60 Gy) and four patients received chemotherapy.
control rates are 89, 84 and 70%, respectively. After a There were no early or late complications due to
median follow-up of 20 months (6–78 months) the brachytherapy or EBRT. The local control and 2-year
6 year OS and DFS are 36 and 27%, respectively. OS and cause-specific survival were 82, 45 and 56%,
The MSKCC treated 322 patients considered respectively (Nori et al. 1995b).
unresectable at thoracotomy and treated with brachy-
therapy (Hilaris and Nori 1987). Patients without
mediastinal nodes metastases achieved 71% local 16 Superior Sulcus Tumors (SST)
control versus 63% in patients with affected mediasti-
nal nodes. The 2 and 3 year OS in N0 and N2 patients The Erasmus Medical Cancer Center Experience in
were 20/15% and 10/3%, respectively. A subgroup of SST has been recently reported (van Geel et al. 2003).
100 patients with positive mediastinal nodes were Twenty-six patients with cytologically or histologi-
treated with surgical resection when feasible, brachy- cally proven NSCLC (T3N0-1 or T4N0) arising in the
therapy (temporary Ir-192 implantation in patients with pulmonary apex were treated with preoperative EBRT
close or positive margins or I-125 implantation in (46 Gy in 23 fractions, 2 Gy per fraction, 5 fractions
patients with residual gross disease) and postoperative per week), surgery and HDR-IORT using a flexible
EBRT (median dose 40 Gy). There was no postoper- intraoperative template (FIT). FIT is a 5 mm thick
ative mortality and local control obtained in 76% of silicone mold in which after loader catheters are
patients (77% for patients with no residual disease and inserted parallel to each other at a fixed distance of
72% in patients who had incomplete or no resection) 1 cm and is used to deliver a homogeneous dose to a
(Hilaris et al. 1983, 1985). surface to which the shape of the mold is adjusted.
Intraoperative Radiotherapy in Lung Cancer 473

Table 4 LDR- HDR-IORT international clinical experiences in stage I–II NSCLC


Authors (reference) Number of Stage Treatment protocol Local control Time point
patients
Hilaris and Mastoras 55 T1-2N0- S ? I-125 (160 Gy) 100% 32% 5 year
(1998) 1 ± (T1N0) OS
EBRT 70% (T2N0)
71% (T1-
2N1)
Fleischman et al. (1992) 14 T1N0 S ? I-125 10/14 (71%) MS 15.1 m
(80–200 Gy)
Fernando et al. (2005) 291 T1N0 Lobar resection (LR) 96.5% (LR)a MS
versus 95.6% (SR)a 68.7 m (LR)a
Sublobar resection (SR) ± I- 50.6 m (SR)a
125
(100–120 Gy)
Lee et al. (2003) 33 T1-2N0 Limited resection 31/33 (94%) 5 year OS
+ 67% (T1N0)
I-125 39% (T2N0)
Voynov et al. (2005) 110 T1-2N0 Limited resection 106/110 5 year OS
+ (96%) 22% (T1N0)
I-125 (100–120 Gy) 5-y LC 90% 12% (T2N0)
S surgery, EBRT external beam radiation therapy, MS median survival, OS overall survival
a
Local recurrence and survival rates for the 2–3 cm tumors

A single radiation fraction of 10 Gy was administered patients who had received no preoperative EBRT or
specified in a plane parallel to the surface of the FIT when the implant presented unacceptable dose dis-
at 1 cm distance with HDR Ir-192. EBRT tribution requirements. The authors describe a 0.8%
(12 9 2 Gy) was indicated for unresectable tumors of postoperative deaths and 17 patients (13%) pre-
during thoracotomy. Three patients progressed during sented nonfatal complications including wound
the preoperative treatment and were excluded. In two infection, empyema with or without broncho-pleural
patients HDR-IORT was not considered because the fistula, bleeding, atelectasia or pneumonia and phle-
tumors had no chest wall invasion. Finally, 21 bitis. The 5 year OS was 25% and patients with
patients underwent the entire programmed treatment negative mediastinal nodes fared better than patients
protocol. One patient (4%) died in the postoperative with positive mediastinal nodes showing a 5 year OS
period due to a cardiac failure. Another patient died of 29 and 10%, respectively.
7 weeks after surgery with a broncho-pleural fistula A summary of NSCLC LDR-IORT and HDR-
and sepsis. Two patients had a prolonged hospital stay IORT clinical experiences from different centers is
of more than 3 weeks because of ARDS and pleural presented in Tables 4 and 5.
empyema recovering after intensive conservative
treatment. With a median follow-up of 18 months, 8
patients were alive (37%), of which 7 had no evidence 17 Summary and Final Considerations
of disease and 18 patients (85%) were free from
locoregional relapse. The median survival for patients The modern developments in the treatment of local-
without and with distant failure was 14 months and ized NSCLC confirm the oncology tendency to
6 months, respectively. intensify systemic and local treatment to promote
Hilaris et al. (1974, 1987) presented the results of disease control. Although a large number of patients
129 patients with SST treated with thoracotomy (in with stage III NSCLC die of systemic disease, local
bloc excision of the involved lung and chest wall failure remains a substantial problem. CALGB
when feasible) interstitial IORT using either perma- reported patterns of disease failure in stage IIIA
nent implantation of I-125 seeds or temporary patients treated with induction chemotherapy, surgery
implantation of Ir-192, and postoperative EBRT in and thoracic irradiation (Kumar et al. 1996). The
474 F. A. Calvo et al.

Table 5 LDR- HDR-IORT international clinical experiences in stage III NSCLC


Authors Number of Stage Treatment protocol Local Time point
(reference) patients control
Valero et al. 20 III S ? PHDRB (16–40 Gy) or IOBT (10.12.5 Gy) 89% 36% 6 year
(2007) or I-125/Pd-103 seeds (90–110 Gy) OS
Burt et al. 225 III S ± I-125a/Ir-192 10/14 MS 12 mb
(1987) (71%) 22% 5 year
OSb
Hilaris and 322 Unresectable Thoracothomy ? I-125 (160 Gy) 71%(N0) 15% 3 year
Nori (1987) 63% OS (N0)
(N2) 3% 3 year
OS (N +)
Hilaris et al. 100 IIIN2 I-125 (160 Gy)/Ir-192 (30 Gy) 89% 22% 5 year
(1985) ± S ? EBRT (30-40 Gy) (R0) OS
53% 22% 5 year
(R1) OS (R2)
72%
(R2)
Nori et al. 12 III ± EBRT (45–60 Gy) + S 82% 45% 2 year
(1995a) (PSM) + OS
I-125/Pd-103 Gelfoam implant
± EBRT (45–60 Gy)
S surgery, EBRT external beam radiation therapy, MS median survival, OS overall survival PHDRB Perioperative high-dose-rate
brachytherapy, IOBT Intra-operative brachytherapy using a silicone mold in which plastic catheters are inserted, PSM Gross o
microscopic positive surgical margins
a
125-I in patients with incomplete resections
b
Patients with incomplete resection and brachytherapy

study found that 52 out of 74 patients had failures and surgical exploration of NSCLC patients. IORT doses
the thorax was the first site of isolated or combined between 10 and 15 Gy combined with EBRT
local failure in 36 patients (69%). (46–50 Gy) induce acute and late toxic events at a
Unfortunately, less than 20% of stage III patients clinically acceptable level. Tables 3, 4 and 5 show
have disease that is resectable for cure at diagnosis summarized international IORT clinical trials
and the optimal management of patients with regarding local control and survival data in NSCLC.
unresectable disease remains controversial. In spite Definitive conclusions based on the available
of improvement in resectability rates with neoad- experiences discussed in this chapter cannot be
juvant approaches, stage III NSCLC patients have a established. In stage I or II NSCLC, IOERT and
high incidence of local recurrence. Based on these IOBT have been used for medically inoperable
observations, higher tumor doses may result in patients with excellent rates of local control
improved local control, and several trials have (70–100%). Alternatively, stereotactic body radio-
emerged in an attempt to promote thoracic control therapy (SBRT) has emerged as a well-tolerated
by escalating total radiation doses exploring altered technique in this subgroup of patients with high rates
fractionation or three-dimensional radiation planning of local control (Fakiris et al. 2009; Lo et al. 2008).
(Rengan et al. 2004; Bradley et al. 2005; Sura et al. IOBT may be reserved to complex central T1-2
2008). tumors or unsuspected surgical findings.
IORT/IOBT has been integrated into the multi- Thoracic control seems to be related to tumor stage
disciplinary management of NSCLC in several small and location, surgical residue and neoadjuvant treat-
prospective single-institution pilot trials as a sophis- ment in locally-advanced NSCLC. Remarkable local
ticated electron, LDR or HDR boost of radiation, control rates in Pancoast’s and stage IIIA tumors with
confirming the feasibility of IORT procedure during microscopic residual disease have been detected.
Intraoperative Radiotherapy in Lung Cancer 475

The effect of IORT on the group of patients pre- Calvo FA, Ortiz de Urbina D, Abuchaibe O et al (1990)
senting with stage IIIB appear to be favorable. This Intraoperative radiotherapy during lung cancer surgery:
technical description and early clinical results. Int J Radiat
point is illustrated by the fact that patients with Oncol Biol Phys 19:103–109
macroscopic residual disease or unresected disease Calvo FA, Santos M, Ortiz de Urbina D (1991) Intraoperative
achieved modest rates of local control (23%), but a radiotherapy in thoracic tumors. Front Radiat Ther Oncol
few long-term survivors are identified. The high rates 25:307–316
Calvo FA, Ortiz de Urbina D, Herreros J, and Llorens R (1992)
of metastatic disease in locally-advanced NSCLC Lung cancer, In: Calvo FA, Santos M, Brady LW (eds)
may conceal the definitive long-term local control but Intraoperative Radiotherapy. Clinical Experiences and
the introduction of novel systemic agents generating results. Springer-Verlag Berlin Heidelberg pp 43–50
more long-term survivors will clarify this question. Calvo FA, Aristu JJ, Moreno M et al (1999) Intraoperative
radiotherapy for lung cancer. In: Van Houte P (ed) Progress
Further confirmatory trials will be necessary to define and perspectives in the treatment of lung cancer. Springer,
the implication of IORT/IOBT in thoracic control and Berlin 173–182
survival of patients with NSCLC. IORT/IOBT as a Cañon R, Azinovic I, Ramis B et al (2008) Intraoperative
component of treatment can be integrated in phase III radiation therapy using mobile linear accelerator in the
multimodality approach to lung cancer. Rev Cancer
trials with treatment strategies that may include surgical (Madrid) 22:27
thoracic exploration. This effort will require interna- Carter YM, Jablons DM, DuBois JB et al (2003) Intraoperative
tional cooperation among expert IORT institutions. radiation therapy in the multimodality approach to upper
aerodigestive tract cancer. Surg Oncol Clin N Am 12:
1043–1063
Chen A, Galloway M, Landreneau R et al (1999) Intraoperative
References 125I brachytherapy for high-risk stage I non-small-cell lung
carcinoma. Int J Radiat Oncol Biol Phys 44:1057–1063
d’Amato TA, Galloway M, Szydlowski G et al (1998)
Abe M, Takahashi M (1981) Intraoperative radiotherapy: the Intraoperative brachytherapy following thoracoscopic
Japanese experience. Int J Radiat Oncol Biol Phys 7(7): wedge resection of stage I lung cancer. Chest 114:
863–868 1112–1115
Arian-Schad Juellner FM, Ratzenhofer B et al (1990) Intraop- De Boer WJ, Mehta DM, Oosterhius JW et al (1989) Tolerance
erative plus external beam irradiation in nonresectable lung of mediastinal structures to intraoperative radiotherapy after
cancer: assessment of local response and therapy-relate side pneumonectomy in dogs. Strahlenther Oncol 165:768
effects. Radiother Oncol 119:137–144 Dong-Soo L, Yeon-Sil K, Jin-Hyoung K, Sang-Nam L, Young-
Aristu J, Rebollo J, Martínez-Monge R, Aramendía JM et al Kyoun K, Myung-Im A et al (2011) Clinical responses and
(1997) Cisplatin, mitomycin, and vindesine followed by prognostic indicators of concurrent chemoradiation for non-
intraoperative and postoperative radiotherapy for stage III small-cell lung cancer. Cancer Res Treat 43(1):32–41
non-small-cell lung cancer: final results of a phase II study. Fakiris AJ, McGarry RC, Yiannoutsos CT et al (2009)
Am J Clin Oncol 20:276–281 Stereotactic body radiation therapy for early-stage non-
Aristu JJ, Calvo FA, Martínez R, Dubois JB, Santos M, Fisher S, small-cell lung carcinoma: four-year results of a prospective
Azinovic I (1999) Lung cancer: EBRT with or without IORT. phase II study. Int J Radiat Oncol Biol Phys 75(3):677–682
In: Gunderson LL, Willet CG, Harrison LB, Calvo FA, Fernando HC, Santos RS, Benfield JR et al (2005) Lobar and
directores. Intraoperative irradiation. Techniques and results. sublobar resection with and without brachytherapy for small
Totowa: Humana Press pp 437–453 stage IA non-small-cell lung cancer. J Thorac Cardiovasc
Barnes M, Pass H, De Luca A et al (1987) Response of Surg 129:261–267
mediastinal and thoracic viscera of the dog to intraoperative Fisher S, Fallahnejad M, Lisker S et al (1994) Role of
radiation therapy (IOERT). Int J Radiat Oncol Biol Phys intraoperative radiation therapy (IORT) for stage III non-
13:371–378 small-cell lung cancer. Hepato-gastroenterol 41:15
Biggs P, Noyes D, Willett C (2003) Clinical physics, Fleischman EH, Kagan AR, Streeter OE et al (1992) Iodine125
applicators choice, technique and equipment for electron interstitial brachytherapy in the treatment of carcinoma of
intraoperative radiation therapy. Surg Oncol Clin N Am the lung. J Surg Oncol 49:25–28
12:899–924 Hansen EK, Roach M III (2010) Handbook of evidence-based
Bradley J, Graham MV, Winter K, Purdy JA, Komaki R, Roa radiation oncology, 2nd edn. Springer, New York, Heidel-
WH et al (2005) Toxicity and outcome results of ROG berg Dordrecht, London
9311: a phase I–II dose-escalation study using three- Hilaris BS, Gomez J, Nori D et al (1985) Combined surgery,
dimensional conformal radiotherapy in patients with inop- intraoperative brachytherapy, and postoperative external
erable non-small-cell lung carcinoma. Int J Radiat Oncol radiation in stage III non-small-cell lung cancer. Cancer
Biol Phys 61:318–328 55:1226–1231
Burt ME, Pomerantz AH, Bains MS et al (1987) Results of Hilaris BS, Mastoras DA (1998) Contemporary brachytherapy
surgical treatment of stage III lung cancer invading the approaches in non-small-cell lung cancer. J Surg Oncol
mediastinum. Surg Clin North Am 67:987–1000 69:258–264
476 F. A. Calvo et al.

Hilaris BS, Martini N, Luomanen RK et al (1974) The value of Merrick HW, Dobelbower RR (2003) Intraoperative radiation
preoperative radiation therapy in apical cancer of the lung. therapy in surgical oncology. Surg Oncol Clin N Am 12:
Surg Clin North Am 54:831–840 883–897
Hilaris BS, Martini N, Wong GY et al (1987) Treatment of Nori D, Li X, Pugkhem T (1995a) Intraoperative brachytherapy
superior sulcus tumor (Pancoast tumor). Surg Clin North using Gelfoam radioactive plaque implants for resected
Am 67:965–977 stage III non-small-cell lung cancer with positive margin: a
Hilaris BS, Nori D, Beattie EJ Jr et al (1983) Value of pilot study. J Surg Oncol 60:257–261
perioperative brachytherapy in the management of non-oat Nori D, Li X, Pugkhem T (1995b) Intraoperative brachytherapy
cell carcinoma of the lung. Int J Radiat Oncol Biol Phys using Gelfoam radioactive plaque implants for resected
9:1161–1166 stage III non-small-cell lung cancer with positive margin: a
Hilaris BS, Nori D (1987) The role of external radiation and pilot study. J Surg Oncol 60:257–261
brachytherapy in unresectable non-small-cell lung cancer. Pass HI, sindelar WF, Kinsella TJ et al (1987) Delivery of
Surg Clin North Am 67:1061–1071 intraoperative radiation therapy after pneumonectomy:
ISIORT0 98 (1998) In: Proceedings of the 1st congress of the experimental observations and early clinical results. Ann
international society of intraoperative radiation therapy. 6–9 Thorac Surg 44:14–20
Sep, Pamplona, España, Rev Med Univ Navarra vol XLII: n Ramalingam SS, Owonikoko TK, Khuri FR (2011) Lung
extraordinario pp 13–68 cancer: new biological insights and recent therapeutic
Jakse G, Kapp KS, Geyer E et al (2007) IORT and external advances. CA Cancer J Clin 61:91–112
beam irradiation (EBI) in clinical stage I-II NSCLC Rengan R, Rosenzweig KE, Venkatraman E, Koutcher LA, Fox
patients with severely compromised pulmonary function: JL, Nayak R et al (2004) Improved local control with higher
an 52-patient single-institutional experience. Strahlenther doses of radiation in large-volume stage III non-small-cell
Onkol 183(2):24–25 lung cancer. Int J Radiat Oncol Biol Phys 60:741–747
Jemal A, Center MM, DeSantis C, Ward EM (2010) Global Sindelar WF, Hoekstra HJ, Kinsella TJ et al (1992) Response of
patterns of cancer incidence and mortality rates and trends. the canine esophagus to intraoperative electron beam
Cancer Epidemiol Biomarkers Prev 19:1893–1907 radiotherapy. Int J Radiat Oncol Biol Phys 25:663–669
Jeuttner FM, Arian-Schad K, Porsch G et al (1990) Intraoper- Sura S, Greco C, Gelblum D, Yorke ED, Jackson A, Rosenzweig
ative radiation therapy combined with external irradiation in KE (2008) 18F-fluorodeoxyglucose positron emission
non resectable non-small-cell lung cancer: preliminary tomography-based Assessment of local failure patterns in
report. Int J Radiat Oncol Biol Phys 18:1143–1150 non-small-cell lung Cancer treated with definitive radiother-
Juettner Smolle, Geyer E, Kapp KS et al (1994) Evaluating apy. Int J Radiat Oncol Biol Phys 70:1397–1402
intraoperative radiation therapy (IORT) and external beam Tochner ZA, Pass HI, Sindelar WF et al (1992) Long term
radiation therapy (EBRT) in non-small-cell lung cancer tolerance of thoracic organs to intraoperative radiotherapy.
(NSCLC). Eur J Cardio-thorac Surg 8:511–516 Int J Radiat Oncol Biol Phys 22(1):65–69
Kritskaia NG, Dobrodeev AI, Zav’ialov AA et al (2006) Valero J, Martinez-Monge R, Pagola M et al (2007) Rescate
Morphofunctional changes in the bronchial epithelium in quirúrgico con técnicas de braquiterapia intraoperatoria en
combined therapy for lung cancer. Arkh Patol 68:10–14 cáncer de pulmón con enfermedad residual tras tratamiento
Kumar P, Herndon J, Langer M, Kohman LJ, Elias AD, Kass quimiorradioterápico o con enfermedad recurrente tras
FC et al (1996) Patterns of disease failure after trimodality radioterapia previa. Clin Transl Oncol 9(Ext 3):5 (a618)
therapy of non-small-cell lung carcinoma pathologic stage van Geel AN, Jansen PP, van Klaveren RJ et al (2003) High
IIIA (N2). Analysis of cancer and leukemia group b relapse-free survival after preoperative and intraoperative
protocol 8935. Cancer 77(11):2393–2399 radiotherapy and resection for sulcus superior tumors. Chest
Lee W, Daly BD, DiPetrillo TA et al (2003) Limited resection 124:1841–1846
for non-small-cell lung cancer: observed local control with Voynov G, Heron DE, Lin CJ et al (2005) Intraoperative (125)I
implantation of I-125 brachytherapy seeds. Ann Thorac Vicryl mesh brachytherapy after sublobar resection for
Surg 75:237–242 high-risk stage I non-small-cell lung cancer. Brachytherapy
Lo SS, Fakiris AJ, Papiez L et al (2008) Stereotactic body 4:278–285
radiation therapy for early-stage non-small-cell lung cancer. Zhou GX, Zeng DW, and Li WH (1992) Acute responses of the
Expert Rev Anticancer Ther 8:87–98 mediastinal and thoracic viscera of canine to intraoperative
Martínez-Monge R, Herreros J, Aristu JJ, Aramendía JM, irradiation. In: Schildberg FW, Wilich N, and Krämling HJ
Azinovic I (1994) Combined treatment in superior sulcus (eds.), Intraoperative radiation therapy, proceedings 4th
tumor. Am J Clin Oncol 17:317–322 internetional symposium, Munich pp 50–52
Brachytherapy for Lung Cancer
A. Polo, M. Castro, A. Montero, and P. Navı́o

Contents Abstract
Brachytherapy is the direct placement of a radio-
1 Introduction.............................................................. 478 active source inside or close to a tumor mass. The
2 Procedure for Lung Brachytherapy...................... 478 use of endobronchial brachytherapy for broncho-
2.1 Endobronchial High-dose Rate Brachytherapy ........ 478 genic carcinoma is not new, being the initial use
2.2 Interstitial Brachytherapy .......................................... 479 reported back in the early 1920s. Brachytherapy
3 Clinical Results: Tumor Control for lung cancer can be done either by implanting
and Palliation ........................................................... 480 the source directly via the upper airway (endolu-
3.1 Endobronchial Brachytherapy (EBB) with Curative minal brachytherapy) or by placing the source
Intention ..................................................................... 480
3.2 EBB with Palliative Intention ................................... 482 interstitially during tumor resection (intraoperative
3.3 Interstitial Brachytherapy .......................................... 484 interstitial brachytherapy) or using a percutaneous
technique (interstitial brachytherapy). Endolumi-
4 Clinical Results: Toxicity........................................ 484
nal high dose rate brachytherapy is largely used for
5 Conclusion ................................................................ 486 the curative and palliative treatment of endobron-
References.......................................................................... 486 chial tumors. Endoluminal brachytherapy can be
used to treat patients with respiratory symptoms
which are predominantly due to the endobronchial
component of their disease. Brachytherapy can
obtain palliation with less morbidity than external
irradiation. Endoluminal brachytherapy can be
used in combination with external beam radiother-
apy for dose escalation as a part of a more radical
approach. Finally, brachytherapy may be given to
patients who require further palliation having
relapsed after previous treatments, including high
dose external beam irradiation. Interstitial brachy-
therapy has been described for the treatment of
A. Polo (&)  A. Montero malignant thoracic tumors. Intraoperative perma-
Department of Radiation Oncology, nent radioactive 125-I seed implantation can be
Ramon y Cajal University Hospital, used in the treatment of lung cancer when resec-
Madrid, Spain
e-mail: alfredo.polo@mac.com tion margins are close or involved with tumor or
for palliation of inoperable disease. Percutaneous
M. Castro  P. Navío
Department of Pneumology, implantation of radioactive seeds has also been
Ramon y Cajal University Hospital, reported for the management of stage T1N0M0
Madrid, Spain

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 477
DOI: 10.1007/174_2011_311, Ó Springer-Verlag Berlin Heidelberg 2011
478 A. Polo et al.

medically inoperable NSCLC with CT-guided bronchoscope. Iodine-125 and gold-198 have also
brachytherapy. During the last two decades, tech- been used for permanent interstitial transbronchial
nological advances helped the development of implantation. These early techniques were not very
brachytherapy and now it is a safe and effective popular due to several logistical reasons, the risk of
standard procedure in the treatment of lung cancer. severe complications, and the poor control over the
dosimetric outcome. The development of the after-
loading techniques during the 1950–1960s was
essential for the widespread application of brachy-
1 Introduction therapy (Henschke et al. 1964). Other new radioactive
sources were developed, such as iodine and cesium.
Brachytherapy is the direct placement of a radioactive New rules of implantation and dose calculation
source inside or close to a tumor mass. This can be were established. These developments introduced
done either by implanting the source directly via the new possibilities for implantations. Afterloading
upper airway (endoluminal brachytherapy) or by techniques were developed to facilitate intraluminal
placing the source interstitially during tumor resection brachytherapy using cesium-137, cobalt-60, or
(intraoperative interstitial brachytherapy) or using a iridium-192. A modern technique for endoluminal
percutaneous technique (interstitial brachytherapy). brachytherapy (enclosing the brachytherapy source
Endoluminal high-dose rate brachytherapy is lar- within a polyethylene catheter and implanting it via a
gely used for the curative and palliative treatment of flexible fiberoptic bronchoscope) was described in the
endobronchial tumors. Endoluminal brachytherapy can 1980s (Moylan et al. 1983; Mendiondo et al. 1983).
be used to treat patients with respiratory symptoms Iridium-192 became the isotope of choice for brach-
which are predominantly due to the endobronchial ytherapy around this time. Although this led to the
component of their disease. Brachytherapy can obtain more frequent use of this treatment, it was still of
palliation with less morbidity than external irradiation. limited application because of the large size and low
Endoluminal brachytherapy can be used in combina- activity of the radioactive sources, prolonged treat-
tion with external beam radiotherapy for dose escala- ment times because of the low-dose rate, and the
tion as a part of a more radical approach. Finally, exposure of staff to significant radiation doses.
brachytherapy may be given to patients who require In addition, both permanent interstitial implantation
further palliation having relapsed after previous treat- and temporary intraluminal brachytherapy required
ments, including high-dose external beam irradiation. general anesthesia for insertion of the sources or
Interstitial brachytherapy has been described for their carrying applicators. During the last two
the treatment of malignant thoracic tumors (Stewart decades, technological advances solved some of the
et al. 2009). Intraoperative permanent radioactive 125I inconveniences of brachytherapy for lung cancer
seed implantation can be used in the treatment of lung (Schray et al. 1988, 1985; Seagren et al. 1985). High
cancer when resection margins are close or involved activity, miniaturized 192Ir sources and computerized
(R1, R2) with tumor or for palliation of inoperable planning and afterloading developed. Intraluminal
disease. Percutaneous implantation of radioactive catheter positioning using local anestesia and flexible
seeds has also been reported by Martinez-Monge bronchoscopy is a safe and effective standard proce-
(2008) for the management of stage T1N0M0 medi- dure (Mehta et al. 1992; Lo et al. 1995).
cally inoperable NSCLC with CT-guided brachy-
therapy. The use of permanent seed brachytherapy has
the potential to improve local control through the 2 Procedure for Lung Brachytherapy
delivery of a high conformal dose. Due to the low
energy of 125I, the falloff in the dose allows optimal 2.1 Endobronchial High-dose Rate
sparing of normal tissues surrounding the implant. Brachytherapy
The initial use of endobronchial brachytherapy for
bronchogenic carcinoma was reported in the early It requires the insertion of one or several plastic
1920s by Yankauer (1922), who implanted radium vectors (usually, a 5–6F closed end plastic tube) into a
capsules into an endobronchial tumor using a rigid patient’s airway. Plastic tubes can be connected to an
Brachytherapy for Lung Cancer 479

afterloader device holding a miniature iridium or codeine pill to the patient before the procedure.
cobalt source able to travel to the desired position Severe acute effects such as massive bleeding or
thanks to a computerized dosimetric system. Per- pneumothorax induction are very rare.
formed by an experienced bronchoscopist, HDR HDR is usually delivered with 1–6 fractions/
brachytherapy has a few acute side effects as routine treatment at an interval of 1–3 weeks. Prescription
fiberoptic bronchoscopy and can therefore be easily doses range between 3 and 20 Gy/fraction (at 0.5–
applied in an outpatient setting. 1 cm from the source axis). In patients previously
This procedure may be performed under intrave- treated with external beam radiation therapy and in
nous conscious sedation typically used in standard the palliative setting, a regimen of 7–10 Gy (HDR)/
bronchoscopic procedures. After topical anesthetic is fraction and a total of 2–3 fractions/treatment can be
carefully applied to the nasal cavity and the naso- used. However, the optimal dosage and fractionation
pharynx, the pulmonologist inserts the bronchoscope schemes for the tumor therapy are still unknown and
through the nose, the vocal cords, and into the the selection of the treatment schedule depends on the
affected bronchus. If there is subtotal stenosis of the approach (palliative, exclusive brachytherapy with
bronchi due to submucosal or exophytic tumor curative intent or boost to external beam radiother-
growth, it is sometimes necessary to perform recan- apy) and the tradition of the team.
alization methods like laser, cryotherapy, or balloon
dilatation for better applicator placement. When the
bronchoscope is in position near the tumor, a thin 2.2 Interstitial Brachytherapy
(5–6 French) polyethylene catheter is inserted through
the bronchoscope working channel. The pulmonolo- The techniques of interstitial implantation vary
gist can see the catheter as it emerges from the distal depending on the type of implantation used. Perma-
end of the bronchoscope and is advanced distal to the nent implants are interstitial implants in which the
tumor area (Gross Tumor Volume—GTV) under radioactive sources are permanently left in the tissue.
direct visualization. Temporary implants are interstitial implants in which
Endoluminal irradiation should be delivered with a the radioactive sources are removed after the desired
‘‘safety margin’’ at both ends of the GTV to account dose has been delivered.
for microscopically disease (Clinical Target Seeds can be placed directly into a tumor as a
Volume—CTV) and catheter movement (Planning volume implant, or mesh in a grid pattern in a planar
Target Volume—PTV). External marks in the cathe- implant (Stewart et al. 2009). Volume implants are
ter’ surface help in the definition of the different often done through a thoracotomy approach. The first
volumes related to the end. As the distal end of the step of the procedure consists of delineating the area
tumor cannot always be seen, the distal endpoint must to be treated (GTV) and the margins to obtain the
be estimated from previous chest X-rays or CT scans CTV. The second step consists of determining the
and controlled during bronchoscopy by fluoroscopy. number of sources required using a previsional
The use of multiple catheters to enlarge the treated dosimetry. Next, hollow needles are inserted, and then
volume and cover complex tumor shapes and local- is the afterloading with radioactive seeds using the
izations (bulkier tumors involving more than one of Mick applicator or preloaded needles.
the bronchi) may be necessary. If additional catheters In the surgical treatment of large tumors, negative
are needed to encompass the tumor, the procedure is margins are often unattainable. In tumors adhering to
repeated. Once the first tube is in place, the bron- critical vessels or bone or other visceral structures,
choscope is then withdrawn, leaving the catheter in resection is often incomplete. To cover the post-
place. The bronchoscope can be again introduced in surgical bed, a planar implant can be custom-made
parallel to check the catheter position and replace it if intraoperatively. The target region is measured and a
necessary. Finally, the catheters are carefully fixed planar implant is prepared. The mesh can be done
and referenced. using permanent vycril suture, gelfoam, or other
Acute side effects of the placement procedure platform custom-sized to cover the region of interest,
include more or less severe coughing, which could be and embedding 125I or 103Pd seeds. The mesh is then
minimized by thorough topical anesthesia and given a sutured to the region of interest to prevent slippage.
480 A. Polo et al.

While permanent seed implantation has been used from EBB as a component of treatment associated
with some success in the intraoperative treatment of with radiotherapy, or as exclusive treatment. Lung
unresectable, residual, or medically inoperable cancers in very early stages, confined to the trachea/
NSCLC its use in a percutaneous CT-guided approach bronchus and without evident lung parenchymal
is merely anecdotal (Martinez-Monge et al. 2008; involvement, the so-called hidden lung carcinomas
Sider et al. 1988; Heelan et al. 1987). This fact may (pure endobronchial carcinoma not visible on CT) are
be explained in part by the lack of tradition even in most likely to benefit from exclusive brachytherapy
the most experienced brachytherapy centers, the small with curative intent. Occasionally, EBB has also been
number of suitable candidates that probably stands for associated with surgery in a postoperative setting as a
less than 20% of those referred for radical radiation boost to the surgical bed in cases of close or micro-
due to a poor surgical risk and the fear of acute scopically affected bronchial resection margins.
complications associated with the procedure. The There are no prospective, randomized studies
implant is performed in a standard CT room under evaluating the effect of endobronchial brachytherapy
general anesthesia, allowing patient immobilization alone in the treatment of very early lung cancer, but
and limited lung motion, decreasing the risk of numerous retrospective studies have reported the
pneumothorax. CT imaging determined the coordi- results of EBB in this context. Table 1 shows the
nates of the skin entry point and permitted adjustment results observed by different groups using exclusively
of depth and angle of needle placement. After target EBB with curative intent (Hilaris et al. 1987;
volume determination, interstitial needles are inserted Schraube et al. 1993; Sutedja et al. 1994; Tredaniel
into the tumor and then seeds are inserted through et al. 1994; Perol et al. 1997; Taulelle et al. 1998;
each needle. Once the implant is completed, a final Stout et al. 2000; Peiffert et al. 2000; Marsiglia et al.
CT scan is obtained for postplanning. 2000; Hennequin et al. 2007; Aumont-le Guilcher
Temporary implants using HDR are useful in the et al. 2011). Nevertheless, it is necessary to keep in
treatment of some clinical situations. A hollow mind the enormous heterogeneity between studies,
stainless needle is passed through the chest skin of the both in the characteristics of the patients enrolled, and
patient. The closed end of a plastic catheter is threa- in terms of the total dose, fractionation scheme, and
ded through the needle until it emerges from the overall treatment time. In spite of this, a benefit of
opposite end of the needle. The needle is then EBB can be inferred from the analysis of these
removed while the plastic tube is held in the chest. studies, mainly in terms of local disease control, that
The process is repeated for the planed number of always must be balanced according to the risk of
afterloading catheters constituting the planar implant. complications.
Each catheter is placed in the desired situation In patients with tumors considered unresectable,
according to the defined target volume and secured brachytherapy has also been used in conjunction with
with suture material. The afterloading catheters are external beam radiation therapy as a boost. Higher
secured to the skin with buttons and silk sutures. combined doses of EBRT and brachytherapy have
Prescription doses ranges between 125 Gy for been associated with increased overall response,
103
Pd and 100–160 Gy for 125I, depending on the although this was not found by all published studies.
approach (adjuvant vs. exclusive), type of implant The observed results are less clear, probably due to
(planar vs. volumetric) and the tradition of the team. the heterogeneity of the series and the different
schemes used. Table 2 lists the clinical outcomes
observed in studies in this field (Aygun et al. 1992;
3 Clinical Results: Tumor Control Cotter et al. 1993; Nori et al. 1993; Speiser and
and Palliation Spratling 1993a; Kohek et al. 1994; Fuwa et al. 2000;
Nomoto et al. 1997; Huber et al. 1997; Furuta et al.
3.1 Endobronchial Brachytherapy (EBB) 1999; Muto et al. 2000; Saito et al. 2000; Langendijk
with Curative Intention et al. 2001; Ozkok et al. 2008). In a prospective,
randomized trial performed by Huber et al. (1997)
Unresectable tumors with a significant intraluminal including a total of 98 patients, two groups were
component and obstructive symptoms can benefit compared. One group was treated with external
Brachytherapy for Lung Cancer 481

Table 1 Definitive brachytherapy alone: clinical results


Author n EBRT (Gy) BT Total dose MFU LC (%) CSS (%) OS (%) MST
Technique (dose per (months)
fraction/
number
of fractions)
Hilaris 55 44% EBRT LDR/HDR 54 63 NA 32 NA
et al. mediastinum
(1987)
Schraube 13 – HDR 5–30 NA NA NA NA 9 months
et al. (3–5/1–6)
(1993)
Sutedja 2 – HDR 30 (10/3) 40 NA 100 NA NA
et al. (1994)
Tredaniel 29 – HDR 42 (7/6) 23 NA NR NA Not reached
et al.
(1994)
Perol et al. 19 – HDR 35 (7/5) 28 75 78 NA 28 months
(1997) (1 years) (1 years)
58
(2 years)
Taulelle 23 – HDR 24–40 32 NA 46 NA 17 months
et al. (8–10/3–4) (2 years)
(1998)
Stout et al. 49 – HDR 15 (15/1) NA NA NA 2 250 days
(2000) 50 30 – – 10 287 days
P = 0.04 P = 0.04
Peiffert 33 No EBRT HDR 30 (5/6) 14 NA 53 80 23 months
et al. (18p) (2 years) (2 years)
(2000) 50–60 (15p) HDR 10–20 (5/2–4)
Marsiglia 34 – HDR 30 (5/6) 29 85 NA 78 NA
et al. (2 years) (2 years)
(2000)
Hennequin 106 – HDR 30–42 (5–7/6) 48 60.3 67.9 47.4 21.4 months
et al. (2 years) (2 years) (2 years)
(2007) 51.6 48.5 24
(5 years) (5 years) (5 years)
Aumont-le 226 – HDR 24–35 30.4 68 81 57 28.6 months
Guilcher (2 years) (2 years) (2 years)
et al. (5–7/4–6) 50 56 29
(2011)
(5 years) (5 years) (5 years)
EBRT: external beam radiotherapy; BT: brachytherapy; HDR: high-dose rate; LDR: low-dose rate; MFU: median follow-up;
LC: local control; CSS: cancer-specific survival; OS: overall survival; MST: median survival time; NA: not available

radiotherapy alone (planned dose 60 Gy), the second Finally, EBB has been also used as adjunctive
group received an additional boost of HDR brachy- treatment to radical surgery. The presence of micro-
therapy (4.8 Gy scheduled, at 10 mm from the source scopic disease in the surgical resection margin of the
axis) before and after external irradiation. In patients bronchial stump is a known risk factor for local
with squamous cell carcinoma, the HDR brachyther- recurrence. The study by McKenna et al. (2008)
apy group showed a borderline advantage in median discusses their experience in the use of EBB as
survival and a better local tumor control. adjuvant treatment after surgery.
482 A. Polo et al.

Table 2 Definitive brachytherapy boost with external beam radiotherapy: clinical results
Author n EBRT BT Total dose MFU LC (%) CSS (%) OS (%) MST
(Gy) Technique (dose per (months)
fraction/
number
of fractions)
Aygun et al. 62 50–60 HDR 15–25 (5/3–5) 18 months NA NA NA 13
(1992)
Cotter et al. 65 55–66 LDR 6–35 NA NA 23 8
(1993) (2 years)
Nori et al. 17 *50 HDR 15 (5/3) 14.5 months 88 NA NA 17.5
(1993) (6 months)
Speiser and 50 60 HDR 22.5–30 NA NA NA NA 11
Spratling (7.5–10/3)
(1993a)
Kohek et al. 39 50–70 HDR 5–25 (5/1–5) NA NA NA NA 13
(1994)
Fuwa et al. 41 50 LDR 22 0– NA NA 61 (CR) NA
(2000) 60 months
Nomoto 9 40–60 HDR 18 (6/3) NA NA NA 64 NA
et al. (1997) (3 years)
Huber et al. 56 60 HDR 9.6 (4.8/2) 2.5 years 12 weeks NA 25 10
(1997) (1 year)
42 60 – – 21 weeks 19 8
(1 year) (P = 0.09)
Furuta et al. 5 40 HDR 18 (6/3) 36 months 100 (CR) 80 (CR) 60 (CR) NA
(1999)
Muto et al. 320 60 HDR 10–15 5– NA NA NA 11
(2000) (5–10/1–3) 36 months
Saito et al. 64 40 LDR 25 44 months 87 96 72 NA
(2000) (5 years) (5 years) (5 years)
Langendijk 48 30–60 HDR 15 (7.5/2) 12 months NA NA 9 (CR) 7
et al. (2001)
47 30–60 – – 15 (CR) 8.5
Ozkok et al. 43 60 HDR 15 (5/3) NA NA NA 25.5 11
(2008) (2 years)
BT and Surgery
McKenna 48 Wedge HDR 24.5 (3.5/7) 13.5 92 (CR) NA 83 (CR) NA
et al. (2008) resection
EBRT: external beam radiotherapy; BT: brachytherapy; HDR: high-dose rate; LDR: low-dose rate; MFU: median follow-up;
LC: local control; CSS: cancer-specific survival; OS: overall survival; MST: median survival time; CR: crude rate; NA: not available

3.2 EBB with Palliative Intention The American Brachytherapy Society (ABS)
should consider EBB as a good palliative treatment in
As mentioned above, one of the main indications for patients with tumors with significant endobronchial
endobronchial brachytherapy is the palliation of component that result in symptoms of shortness of
symptoms due to uncontrolled bronchial tumor breath, persistent cough, hemoptysis, or signs of
growth. EBB is a relatively simple and quick method obstructive pneumonitis (Nag et al. 2001; Gaspar
to relieve bronchial obstruction and ensure the 1998). Patients should be able to withstand the proce-
viability of the airway for a long time. dure, should not present a significant active bleeding,
Brachytherapy for Lung Cancer 483

Table 3 Palliative brachytherapy: clinical results


Author n Prior BT BT schedule Symptomatic response
treatments technique
Lo et al. (1992) 77 laser LDR 45–60 Gy Overall: 54%
Gollins et al. (1996) 322 EBRT, HDR 15 Gy 9 1 Stridor: 92%;
laser 20 Gy 9 1 hemoptysis 88%;
cough: 62%;
dyspnea: 60%;
pain: 50%;
pulmonary collapse: 46%
Speiser and 109 laser HDR 7.5 Gy 9 3 Overall: 70%;
Spratling (1993a) 5 Gy 9 3 hemoptysis: 99%;
pneumonia: 99%;
dyspnea: 86%;
cough: 85%
Delclos et al. (1996) 81 EBRT HDR 15 Gy 9 2 Overall: 84%
Ornadel et al. 117 EBRT, HDR Cough: 62–77%;
(1997) laser dyspnea: 32–56%;
hemoptysis: 78–97%
Stout et al. (2000) 50 None EBRT 30 Gy Improved symptom palliation for EBRT (83%)
(3.75 9 8) versus HDR (59%), P = 0.03
49 HDR 15 Gy 9 1
Kelly et al. (2000) 175 EBRT, HDR 15 Gy 9 1 Overall: 66%
laser
Celebioglu et al. 95 EBRT HDR 7.5 Gy 9 3 Overall: 100%
(2002) 10 Gy 9 2
Mallick et al. (2007) 95 NA HDR 10 Gy 9 1 Dyspnea: 92.5%;
8 Gy 9 2 cough: 81%;
hemoptysis: 97%;
15 Gy 9 1 pneumonia: 91%
Kubaszewska et al. 270 EBRT, BT HDR 8 Gy 9 1 Overall: 80%;
(2008) 10 Gy 9 1 dyspnea: 76%;
cough: 77%;
hemoptysis: 92%;
pneumonia: 82%
Skowronek et al. 648 EBRT HDR 7.5 Gy 9 3 Overall:88%; no differences between schedules
(2009) (303p)
10 Gy 9 1
(345p)
EBRT: external beam radiotherapy; BT: brachytherapy; HDR: high-dose rate; LDR: low-dose rate; NA: not available

and should have tumors that still protruding into the or in combination with other therapies such as EBRT,
bronchial lumen, allow the passage of the endobron- Nd: YAG laser, cryotherapy or photodynamic therapy.
chial catheter. However, EBB is not always indicated Again, the huge disparity of criteria for inclusion in the
in patients with obstructive symptoms. Tumors that studies, the different treatment regimens used and the
cause bronchial luminal narrowing by endobronchial aspects considered when assessing the effect of palli-
growth are more likely to respond than those with an ation obtained, makes it difficult to establish firmly and
extrinsic component and obstruction secondary to definitively the role of EBB. Table 3 shows the results
compression exerted on bronchus or trachea. of symptomatic improvement observed in studies
Multiple studies have analyzed the role of EBB as a involving more than 50 patients using EBB as
palliative treatment of lung cancer, either exclusively palliative treatment (Speiser and Spratling 1993a;
484 A. Polo et al.

Lo et al. 1992; Gollins et al. 1996; Delclos et al. 1996; of this fact, EBB remains a reasonable option in the
Ornadel et al. 1997; Kelly et al. 2000; Celebioglu et al. palliative management of a patient with endobron-
2002; Mallick et al. 2007; Kubaszewska et al. 2008; chial lesion causing obstruction or hemoptysis who
Skowronek et al. 2009). has previously received thoracic EBRT (Rodrigues
To our knowledge, there is only one controlled, et al. 2011).
randomized study to evaluate the effect of dose rate,
overall radiation dose, fractionation, and localization
of the afterloading catheter to survival rate, local 3.3 Interstitial Brachytherapy
control and complications (Huber et al. 1995). In this
study, two treatment regimens with a comparable total Patients are referred for brachytherapy by the surgeon
irradiation dose of 15 Gy (at 1 cm from the source preoperatively when there is a concern for incomplete
axis), but different doses per fraction (four fractions of tumor resection or close or positive margins. In other
3.8 Gy on a weekly basis, and two fractions of 7.2 Gy cases, tumor may be stripped off vital structures, such as
at a 3-week interval) were compared. They found no blood vessels or nerves. On the other hand, in patients
disadvantages for the shorter fractionation regimen. with poor cardiopulmonary reserve, a sublobar resection
Similar survival time (19 weeks) and local control time may be considered, but retrospective data have shown
was achieved in both groups. The complication rate that in these patients the rates of local recurrence is
was also similar with fatal hemorrhage occurring in higher. Addition of interstitial seed implantation can
21% of all patients. result in a better local control. This hypotheses is being
Globally, it is estimated that the symptomatic relief tested in the ACOSOG Z4032 randomized trial, where
of brachytherapy ranges between 38 and 90% of cases patients with tumors\3 cm maximal diameter stage IA
with hemoptysis, between 50 and 66% of cases with or selected stage IB (visceral pleural involvement), node
persistent cough and between 43 and 64% cases with negative NSCLC and not fit for lobar resection are
dyspnea, with an average duration of symptomatic randomized to sublobar resection alone or sublobar
response of 4–6 months after treatment (Marsiglia resection with 125I seeds placed at the suture line.
et al. 2000). Table 4 shows the results observed using interstitial
Two systematic reviews have been published on this brachytherapy in lung cancer.
topic. Ontario Cancer Care clinical guidelines analyzed
29 trials involving the use of EBB with palliative
intentions. The authors concluded that in previously 4 Clinical Results: Toxicity
untreated patients, EBRT alone is more effective than
EBB. In patients who have previously received EBRT, Acute complications of the endoluminal brachyther-
EBB is a good option to consider according to the apy procedure are not more frequent than those
patient’s condition (Ung et al. 2006). Similarly, a occurring during routine diagnostic flexible bron-
Cochrane Review in 2008 reviewed 13 trials, but choscopy. The placement of the catheter and the
without carrying out a meta-analysis due to the wide irradiation procedure are normally well tolerated. The
heterogeneity in terms of patients enrolled and their commonest acute effect which can be attributed to
treatments. The authors concluded that EBRT is more endoluminal brachytherapy is a transient exacerbation
effective than EBB and there is no conclusive evidence of cough, usually within 2–3 weeks after treatment.
for recommending the combination of EBRT and EBB The most important potential side effect of end-
versus EBRT alone (Cardona et al. 2008). oluminal brachytherapy is fatal hemoptysis. Massive
Finally, the American Society for Radiation haemoptysis is usually a fatal event which occurs in
Oncology (ASTRO) has recently published an evi- lung cancer whether or not radiotherapy has been
dence-based clinical practice guideline regarding given. The incidence of this complication ranges
palliative thoracic radiotherapy. The role of EBB is between 0 and 32% of patients treated with a
discussed. The authors recognise that there are no prevalence of approximately 10% (Vergnon et al.
randomized trials assessing the role of EBB in the 2006; Bedwinek et al. 1992). Despite numerous
routine initial palliative management of endobron- attempts, a direct relationship between the occur-
chial obstruction resulting from lung cancer. In spite rence of massive hemoptysis and the type of
Brachytherapy for Lung Cancer 485

Table 4 Interstitial brachytherapy: clinical results


Author n Disease stage BT Technique Total dose LC (%) OS (%)
(AJCC) (Gy)
125
Hilaris and Martini 470 I–III I 160 80 (5 years, 7 (5 years)
(1979) stages I, II)
80 (5 years, stage
III)
125
Hilaris and Martini 88 T1–3 N2 I; HDR 160; 30 76 (2 years) 51 (2 years)
(1988) 225 T3N0 22 (2 years)
125
Ginsberg et al. 102 I–III, close/positive I 160 NA 41 (5 years)
(1994) margins HDR 10–20
Brach et al. (1994) 20 I, tumor \2 cm HDR 10–20 NA 75% (CR,
3–30 months)
125
Chen et al. (1999) 23 I I 100–120 100 (CR) 83 (CR)
125
Santos et al. (2003) 203 IA–B I (101p) 100–120 98 (CR) 60 (4 years)
–(102p) – 81 (CR) 67 (4 years)
125
Lee et al. (2003) 33 I I 125–140 94 (5 years) 47 (5 years)
125
Voynov et al. 118 IA–B I 85–129 87 (5 years) 18 (5 years)
(2005)
Birdas et al. (2006) 167 IB Sublobar 100–120 95.2 (CR) 54 (4 years)
resection ? – 96.8 (CR) 52 (4 years)
125
I (41p)
Lobectomy
(126p)
125
Colonias et al. 145 IA–B I 120 NA 35 (5 years)
(2011)
BT: brachytherapy; HDR: high-dose rate; LC: local control; OS: overall survival; CR: crude rate; NA: not available

implants, tumor location, volume or treatment dose external radiotherapy (30 Gy in eight fractions). The
has not been clearly found. Some authors noted a incidence of massive fatal haemoptysis was the same
higher incidence of bleeding in the treatments of in both arms of the trial, occurring in only 7% of the
tumors that settled in the bronchus of right upper patients overall (Stout et al. 2000).
lobe against the left upper lobe, connecting this to Other complications related to endoluminal brach-
the proximity of the brachytherapy catheter to the ytherapy are pulmonary bronchitis in 4–12% of patients
pulmonary artery (Aygun and Blum 1995). Some- (Speiser and Spratling 1993b; Gauwitz et al. 1992) and
times the occurrence of fatal hemoptysis is related to bronchial stenosis in 11% (Speiser and Spratling
the disease progression, and not to the treatment. 1993b). Histological changes consist of mild mucosal
Gollins et al. (1996) reported 8% massive fatal inflammation to severe bronchial fibrosis. Speiser and
haemoptysis in a large retrospective series of 406 Spratling (1993b) have proposed a grading system from
patients. Associated treatment factors increasing the grade 1 (mild mucosal inflammation, thin, circumfer-
likelihood of massive fatal haemoptysis were a ential membrane, no significant luminal obstruction) to
brachytherapy dose greater than 15 Gy, prior laser grade 4 (greater degree of fibrosis with circumferential
treatment, brachytherapy used for reirradiation and stenosis). Therapy of these postradiation effects
concurrent external beam radiotherapy. Stout et al. consists of conventional treatment, including admin-
reported a UK randomised trial where brachytherapy istration of steroids, oxygen, balloon dilatation, laser
(15 Gy) as a sole primary treatment was compared resection, or endobronchial prosthesis placement in
directly with a palliative course of fractionated case of severe stenosis (Vergnon et al. 2006).
486 A. Polo et al.

Overall, endobronchial brachytherapy must be Brach B, Buhler C, Hayman MH, Joyner LRJ, Liprie SF (1994)
considered a well tolerated, with a gentle toxicity Percutaneous computed tomography-guided fine needle brach-
ytherapy of pulmonary malignancies. Chest 106:268–274
profile. It is a not very aggressive treatment option, Cardona AF, Reveiz L, Ospina EG, Ospina V, Yepes A (2008)
especially in patients with poor performance status. Palliative endobronchial brachytherapy for non-small cell
lung cancer. Cochrane Database Syst Rev. :CD004284
Celebioglu B, Gurkan OU, Erdogan S et al (2002) High dose
5 Conclusion rate endobronchial brachytherapy effectively palliates
symptoms due to inoperable lung cancer. Jpn J Clin Oncol
32:443–448
Endoluminal HDR brachytherapy with Iridium-192 Chen A, Galloway M, Landreneau R et al (1999) Intraoperative
has an established role in the treatment of lung cancer 125I brachytherapy for high-risk stage I non-small cell lung
when used alone or with external irradiation as carcinoma. Int J Radiat Oncol Biol Phys 44:1057–1063
described above. Symptomatic improvement can be Colonias A, Betler J, Trombetta M et al (2011) Mature follow-up
for high-risk stage I non-small-cell lung carcinoma treated
achieved in a large proportion of patients, and with sublobar resection and intraoperative iodine-125 brach-
sometimes brachytherapy alone can cure small ytherapy. Int J Radiat Oncol Biol Phys 79:105–109
tumors. Brachytherapy can be combined with all other Cotter GW, Lariscy C, Ellingwood KE, Herbert D (1993)
modalities of tumor therapy, including external beam Inoperable endobronchial obstructing lung cancer treated
with combined endobronchial and external beam irradia-
radiotherapy, local therapies (cryotherapy, photody- tion: a dosimetric analysis. Int J Radiat Oncol Biol Phys
namic therapy, laser resection) and chemotherapy. 27:531–535
However, the optimal dosage and fractionation Delclos ME, Komaki R, Morice RC, Allen PK, Davis M,
schemes for the tumor therapy still have to be defined. Garden A (1996) Endobronchial brachytherapy with high-
dose-rate remote afterloading for recurrent endobronchial
Furthermore, the risk of severe complications could lesions. Radiology 201:279–282
perhaps be avoided through better scheduling and Furuta M, Tsukiyama I, Ohno T et al (1999) Radiation therapy
dosing of the HDR brachytherapy. Rigorous appraisal for roentogenographically occult lung cancer by external
in controlled clinical trials is needed to document the beam irradiation and endobronchial high dose rate brach-
ytherapy. Lung Cancer 25:183–189
potential benefits and identify the risks when multi- Fuwa N, Ito Y, Matsumoto A, Morita K (2000) The treatment
modality treatment is employed. Endoluminal brach- results of 40 patients with localized endobronchial cancer
ytherapy is a permanent exercise of multidisciplinary with external beam irradiation and intraluminal irradiation
venture, and there is a need for close contact between using low dose rate (192)Ir thin wires with a new catheter.
Radiother Oncol 56:189–195
radiation oncologists and chest specialists. Gaspar LE (1998) Brachytherapy in lung cancer. J Surg Oncol
67:60–70
Gauwitz M, Ellerbroek N, Komaki R et al (1992) High dose
endobronchial irradiation in recurrent bronchogenic carci-
References noma. Int J Radiat Oncol Biol Phys 23:397–400
Ginsberg RJ, Martini N, Zaman M et al (1994) Influence of
surgical resection and brachytherapy in the management of
Aumont-le Guilcher M, Prevost B, Sunyach MP et al (2011) superior sulcus tumor. Ann Thorac Surg 57:1440–1445
High-dose-rate brachytherapy for non-small-cell lung Gollins SW, Ryder WD, Burt PA, Barber PV, Stout R (1996)
carcinoma: a retrospective study of 226 patients. Int J Massive haemoptysis death and other morbidity associated
Radiat Oncol Biol Phys 79:1112–1116 with high dose rate intraluminal radiotherapy for carcinoma
Aygun C, Blum JE (1995) Treatment of unresectable lung of the bronchus. Radiother Oncol 39:105–116
cancer with brachytherapy. World J Surg 19:823–827 Heelan RT, Hilaris BS, Anderson LL et al (1987) Lung tumors:
Aygun C, Weiner S, Scariato A, Spearman D, Stark L (1992) percutaneous implantation of I-125 sources with CT treat-
Treatment of non-small cell lung cancer with external beam ment planning. Radiology 164:735–740
radiotherapy and high dose rate brachytherapy. Int J Radiat Hennequin C, Bleichner O, Tredaniel J et al (2007) Long-term
Oncol Biol Phys 23:127–132 results of endobronchial brachytherapy: A curative treat-
Bedwinek J, Petty A, Bruton C, Sofield J, Lee L (1992) The use ment? Int J Radiat Oncol Biol Phys 67:425–430
of high dose rate endobronchial brachytherapy to palliate Henschke UK, Hilaris BS, Mahan GD (1964) Remote afterload-
symptomatic endobronchial recurrence of previously irra- ing with intracavitary applicators. Radiology 83:344–345
diated bronchogenic carcinoma. Int J Radiat Oncol Biol Hilaris BS, Martini N (1979) Interstitial brachytherapy in
Phys 22:23–30 cancer of the lung: a 20 year experience. Int J Radiat Oncol
Birdas TJ, Koehler RP, Colonias A et al (2006) Sublobar Biol Phys 5:1951–1956
resection with brachytherapy versus lobectomy for stage Ib Hilaris BS, Martini N (1988) The current state of intraoperative
nonsmall cell lung cancer. Ann Thorac Surg 81:434–438 interstitial brachytherapy in lung cancer. Int J Radiat Oncol
discussion 438–439 Biol Phys 15:1347–1354
Brachytherapy for Lung Cancer 487

Hilaris BS, Martini N, Wong GY, Nori D (1987) Treatment of brachytherapy of recurrent bronchogenic carcinoma: a
superior sulcus tumor (Pancoast tumor). Surg Clin North new approach using the flexible fiberoptic bronchoscope.
Am 67:965–977 Radiology 147:253–254
Huber RM, Fischer R, Hautmann H et al (1995) Palliative Muto P, Ravo V, Panelli G, Liguori G, Fraioli G (2000) High-
endobronchial brachytherapy for central lung tumors. A dose rate brachytherapy of bronchial cancer: treatment
prospective, randomized comparison of two fractionation optimization using three schemes of therapy. Oncologist
schedules. Chest 107:463–470 5:209–214
Huber RM, Fischer R, Hautmann H, Pollinger B, Haussinger K, Nag S, Kelly JF, Horton JL, Komaki R, Nori D (2001)
Wendt T (1997) Does additional brachytherapy improve the Brachytherapy for carcinoma of the lung. Oncology (Will-
effect of external irradiation? A prospective, randomized iston Park) 15:371–381
study in central lung tumors. Int J Radiat Oncol Biol Phys Nomoto Y, Shouji K, Toyota S et al (1997) High dose rate
38:533–540 endobronchial brachytherapy using a new applicator.
Kelly JF, Delclos ME, Morice RC, Huaringa A, Allen PK, Radiother Oncol 45:33–37
Komaki R (2000) High-dose-rate endobronchial brachy- Nori D, Allison R, Kaplan B, Samala E, Osian A, Karbowitz S
therapy effectively palliates symptoms due to airway (1993) High dose-rate intraluminal irradiation in bronchogenic
tumors: the 10-year M. D. Anderson cancer center experi- carcinoma. Technique and results. Chest 104:1006–1011
ence. Int J Radiat Oncol Biol Phys 48:697–702 Ornadel D, Duchesne G, Wall P, Ng A, Hetzel M (1997)
Kohek PH, Pakisch B, Glanzer H (1994) Intraluminal irradi- Defining the roles of high dose rate endobronchial brach-
ation in the treatment of malignant airway obstruction. Eur J ytherapy and laser resection for recurrent bronchial malig-
Surg Oncol 20:674–680 nancy. Lung Cancer 16:203–213
Kubaszewska M, Skowronek J, Chichel A, Kanikowski M Ozkok S, Karakoyun-Celik O, Goksel T et al (2008) High dose
(2008) The use of high dose rate endobronchial brachy- rate endobronchial brachytherapy in the management of
therapy to palliate symptomatic recurrence of previously lung cancer: response and toxicity evaluation in 158
irriadiated lung cancer. Neoplasma 55:239–245 patients. Lung Cancer 62:326–333
Langendijk H, de Jong J, Tjwa M et al (2001) External Peiffert D, Spaeth D, Menard O, Winnefeld J (2000) High dose
irradiation versus external irradiation plus endobronchial endobronchial brachytherapy: a curative treatment. Cancer
brachytherapy in inoperable non-small cell lung cancer: a Radiother 4:197–201
prospective randomized study. Radiother Oncol 58:257–268 Perol M, Caliandro R, Pommier P et al (1997) Curative
Lee W, Daly BD, DiPetrillo TA et al (2003) Limited resection irradiation of limited endobronchial carcinomas with high-
for non-small cell lung cancer: observed local control with dose rate brachytherapy. Results of a pilot study. Chest
implantation of I-125 brachytherapy seeds. Ann Thorac 111:1417–1423
Surg 75:237–242 discussion 242-3 Rodrigues G, Videtic GM, Sur R (2011) Palliative thoracic
Lo TC, Beamis JFJ, Weinstein RS et al (1992) Intraluminal radiotherapy in lung cancer: an American Society for
low-dose rate brachytherapy for malignant endobronchial Radiation Oncology evidence-based clinical practice guide-
obstruction. Radiother Oncol 23:16–20 line. Pract Radiat Oncol :60–71
Lo TC, Girshovich L, Healey GA et al (1995) Low dose rate Saito M, Yokoyama A, Kurita Y, Uematsu T, Tsukada H,
versus high dose rate intraluminal brachytherapy for malig- Yamanoi T (2000) Treatment of roentgenographically
nant endobronchial tumors. Radiother Oncol 35:193–197 occult endobronchial carcinoma with external beam radio-
Mallick I, Sharma SC, Behera D (2007) Endobronchial therapy and intraluminal low-dose-rate brachytherapy: sec-
brachytherapy for symptom palliation in non-small cell ond report. Int J Radiat Oncol Biol Phys 47:673–680
lung cancer–analysis of symptom response, endoscopic Santos R, Colonias A, Parda D et al (2003) Comparison between
improvement and quality of life. Lung Cancer 55:313–318 sublobar resection and 125Iodine brachytherapy after sublo-
Marsiglia H, Baldeyrou P, Lartigau E et al (2000) High-dose-rate bar resection in high-risk patients with Stage I non-small-cell
brachytherapy as sole modality for early-stage endobronchial lung cancer. Surgery 134:691–697 discussion 697
carcinoma. Int J Radiat Oncol Biol Phys 47:665–672 Schraube P, Fritz P, Becker HD, Wannenmacher M (1993) The
Martinez-Monge R, Pagola M, Vivas I, Lopez-Picazo JM results of the endoluminal high-dose-rate irradiation of
(2008) CT-guided permanent brachytherapy for patients central non-small cell bronchial carcinomas. Strahlenther
with medically inoperable early-stage non-small cell lung Onkol 169:228–234
cancer (NSCLC). Lung Cancer 61:209–213 Schray MF, McDougall JC, Martinez A, Edmundson GK,
McKenna RJJ, Mahtabifard A, Yap J et al (2008) Wedge Cortese DA (1985) Management of malignant airway
resection and brachytherapy for lung cancer in patients with obstruction: clinical and dosimetric considerations using
poor pulmonary function. Ann Thorac Surg 85:S733–S736 an iridium-192 afterloading technique in conjunction with
Mehta M, Petereit D, Chosy L et al (1992) Sequential the neodymium-YAG laser. Int J Radiat Oncol Biol Phys
comparison of low dose rate and hyperfractionated high 11:403–409
dose rate endobronchial radiation for malignant airway Schray MF, McDougall JC, Martinez A, Cortese DA, Brutinel
occlusion. Int J Radiat Oncol Biol Phys 23:133–139 WM (1988) Management of malignant airway compromise
Mendiondo OA, Dillon M, Beach LJ (1983) Endobronchial with laser and low dose rate brachytherapy. The Mayo
brachytherapy in the treatment of recurrent bronchogenic Clinic experience. Chest 93:264–269
carcinoma. Int J Radiat Oncol Biol Phys 9:579–582 Seagren SL, Harrell JH, Horn RA (1985) High dose rate
Moylan D, Strubler K, Unal A, Mohiuddin M, Giampetro A, intraluminal irradiation in recurrent endobronchial carci-
Boon R (1983) Work in progress. Transbronchial noma. Chest 88:810–814
488 A. Polo et al.

Sider L, Mittal BB, Nemcek AAJ, Bobba VS (1988) CT-guided Sutedja G, Baris G, van Zandwijk N, Postmus PE (1994) High-
placement of iodine-125 seeds for unresectable carcinoma dose rate brachytherapy has a curative potential in patients
of the lung. J Comput Assist Tomogr 12:515–517 with intraluminal squamous cell lung cancer. Respiration
Skowronek J, Kubaszewska M, Kanikowski M, Chichel A, 61:167–168
Mlynarczyk W (2009) HDR endobronchial brachytherapy Taulelle M, Chauvet B, Vincent P et al (1998) High dose rate
(HDRBT) in the management of advanced lung cancer— endobronchial brachytherapy: results and complications in
Comparison of two different dose schedules. Radiother 189 patients. Eur Respir J 11:162–168
Oncol 93:436–440 Tredaniel J, Hennequin C, Zalcman G et al (1994) Prolonged
Speiser BL, Spratling L (1993a) Remote afterloading brachy- survival after high-dose rate endobronchial radiation for
therapy for the local control of endobronchial carcinoma. malignant airway obstruction. Chest 105:767–772
Int J Radiat Oncol Biol Phys 25:579–587 Ung YC, Yu E, Falkson C et al (2006) The role of high-dose-
Speiser BL, Spratling L (1993b) Radiation bronchitis and rate brachytherapy in the palliation of symptoms in patients
stenosis secondary to high dose rate endobronchial irradi- with non-small-cell lung cancer: a systematic review.
ation. Int J Radiat Oncol Biol Phys 25:589–597 Brachytherapy 5:189–202
Stewart AJ, Mutyala S, Holloway CL, Colson YL, Devlin PM Vergnon JM, Huber RM, Moghissi K (2006) Place of
(2009) Intraoperative seed placement for thoracic malig- cryotherapy, brachytherapy and photodynamic therapy in
nancy—a review of technique, indications, and published therapeutic bronchoscopy of lung cancers. Eur Respir J
literature. Brachytherapy 8:63–69 28:200–218
Stout R, Barber P, Burt P et al (2000) Clinical and quality of Voynov G, Heron DE, Lin CJ et al (2005) Intraoperative (125)I
life outcomes in the first United Kingdom randomized trial Vicryl mesh brachytherapy after sublobar resection for
of endobronchial brachytherapy (intraluminal radiotherapy) high-risk stage I non-small cell lung cancer. Brachytherapy
vs. external beam radiotherapy in the palliative treatment of 4:278–285
inoperable non-small cell lung cancer. Radiother Oncol Yankauer S (1922) Two cases of lung tumor treated broncho-
56:323–327 scopically. NY Med J 21:741
Limited-Disease Small-Cell Lung Cancer
Branislav Jeremić, Željko Dobrić, and Francesc Casas

Contents Abstract
About one-third of all patients with small cell lung
1 Introduction.............................................................. 492 cancer present with their disease confined to
2 General Treatment Concepts ................................. 492 thorax. In the last two decades, combined radiation
therapy and chemotherapy have been considered
3 Chemotherapy .......................................................... 493
standard treatment option in this disease. This
4 Thoracic Radiation Therapy .................................. 495 particularly became evident when radical thoracic
5 Conclusions ............................................................... 500 radiation therapy was combined with concurrent
References.......................................................................... 500
platinum-etoposide chemotherapy. Prophylactic
cranial irradiation was also shown to be important
part of the overall treatment approach and is
suggested as standard treatment option in the last
decade. Several questions, however, remained in
focus of clinical investigations in the recent
decades with no clear answers provided. In partic-
ular, investigators embarked on evaluating the
timing of administration of radiation therapy and
chemotherapy in combined modality approach.
Several prospective randomized trials and several
meta-analyses showed that there is an advantage
for early administration of the two modalities,
especially using concurrent approach. While land-
mark Intergroup study clearly showed benefit of
BID fractionation over QD fractionation, two
ongoing studies further built on the question of
optimization of dose and fractionation issue in this
setting. Finally, issue of tumor volumes to be used
during the radiation therapy course remain subop-
timally investigated due to lack of clinical studies
investigating it and, therefore, poor guidelines for
B. Jeremić (&)  Ž. Dobrić daily clinical practice.
Institute of Lung Diseases, Sremska Kamenica, Serbia
e-mail: nebareje@gmail.com
F. Casas
University Clinic, Barcelona, Spain

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 491
DOI: 10.1007/174_2011_253, Ó Springer-Verlag Berlin Heidelberg 2011
492 B. Jeremić et al.

available computer-enabled volumetric analysis. The


1 Introduction most recent staging classification of small-cell lung
cancer (Shepherd et al. 2007) and stage grouping
Small-cell lung cancer is highly aggressive carcinoma differentiated tumors with different prognoses. The
representing approximately 15–20% of all lung can- TNM classification and staging system was recom-
cer cases (Greenlee et al. 2000). It is an entity of lung mended for small-cell lung cancer and stratification
cancer that is biologically and clinically different by stage I–III was also recommended in clinical trials
from non-small-cell lung cancer. The world health of early-stage (i.e., limited) disease.
organization (WHO) classification of lung tumor,
revised in 2004 (Travis et al. 2004) remains the cor-
nerstone for lung cancer nomenclature.
Of all potential risk factors which may cause this 2 General Treatment Concepts
disease, cigarette smoking has long been considered as
the primary risk factor for its occurrence. This occurs in Chemotherapy is the mainstay of the treatment for
[90% cases (Mulshine et al. 1993; Ihde et al. 1993). several decades now. With chemotherapy alone,
Cough, hemoptysis, dyspnoea, hoarseness, and however, intrathoracic failure occurs in up to 80%
dysphagia are the most frequent clinical signs and leading to a median survival of 10–14 months
symptoms. The paraneoplastic syndromes are more (Cohen et al. 1979). Radiation therapy has great
common than in non-small-cell lung cancer. They potential in decreasing locoregional failures and it
occur frequently in a variety of presentations, including was increasingly practiced in the seventies and the
the syndrome of inappropriate antidiuretic hormone, eighties of the last century. However, radiation
ectopic Cushing’s syndrome, and Lambert-Eaton therapy was eventually introduced as a necessary
Myastenic Syndrome. Rare neurologic syndromes can part of the combined modality approach owing to
also occur, such as subacute spinal or peripheral neu- results of two meta-analyses that appeared almost
ropathy, cerebellar ataxia, limbic encephalopathia, and two decades ago
retinal degeneration (Curran 2001). (Pignon et al. 1992; Warde and Payne 1992). Small
More than 4 decades ago, the Veterans Adminis- but significant improvement in 2-year and 3-year
tration Lung Group had proposed dividing all small- survival, averaging 5–7% and an improvement in
cell lung cancer cases into the two-stage system: local control rates in 25% cases with the addition of
limited-disease and extensive disease (Green et al. thoracic radiation therapy was shown. Importantly,
1969). Majority of clinicians and investigators still the widespread use of cisplatin/etoposide regimen,
use it nowadays. The vast majority of patients and its lower toxicity (than that observed with the
(approximately two-thirds) fall into the extensive cyclophosphamide, doxorubicin, vincristin) when
disease category while limited-disease occurs in combined with thoracic radiation therapy, made
approximately one-third of all small-cell lung cancer more effective use of concurrent thoracic radiation
cases. Limited-disease small-cell lung cancer is therapy and platinum-based chemotherapy, which is
defined as the disease confined to the hemithorax of nowadays considered as the standard treatment in
origin along with the involved regional lymph nodes limited-disease small-cell lung cancer. While recent
(hilar and mediastinal), with or without ipsilateral meta-analysis (Auperin et al. 1999) confirmed the
supraclavicular lymph nodes. It can also be consid- necessity for prophylactic cranial irradiation, details
ered as a disease which can be incorporated within a of it will be dealt with in another chapter in this
single, tolerable radiation therapy treatment field, and book.
may include patients with contralateral, mediastinal, A number of questions requesting further studies in
or hilar lymph nodes. What has created a confusion this disease remain. These include, but are not limited
and still does it is the term ‘‘tolerable radiation ther- to optimization of both chemotherapy (choice of
apy treatment field’’. It was not always easy to denote drugs and its schedule/timing/dosing) and thoracic
and compare it between clinicians, especially radia- radiation therapy (timing of thoracic radiation therapy
tion oncologists. This, however, may be more easily and dose/volume/fractionation). Some of them will be
solved nowadays due to normal lung constraints and addressed below.
Limited-Disease Small-Cell Lung Cancer 493

(cisplatin or carboplatin) with nonplatinum-based


3 Chemotherapy regimens and did not reveal any difference in survival
for the compared arms. Subgroups (stage, radiation
Due to its pronounced chemosensitivity, there are therapy use) analysis also showed no difference
many chemotherapeutic agents which achieve (Amarasena et al. 2008). Given these conflicting
response rates of [30% in small-cell lung cancer. results, many consider that the optimal chemotherapy
They include cisplatin, carboplatin, etoposide, cyclo- schedule for small-cell lung cancer has not yet been
phosphamide, doxorubicin, methotrexate, and vin- confirmed. Contrasting these ambiguities are the long-
cristine (Sandler, 2003). While cyclophosphamide/ known facts about favorable toxicity profile of cis-
doxorubicin/vincristin regimen was mostly used in platin/etoposide regimen (Pignon et al. 1992) in
early studies, studies done in the last three decades combination with radiation therapy, no evidence of
started more frequently employing cisplatin/etopo- improvement with any other schedule and the best
side, since it became known as not only less toxic, but results so far obtained with this schedule which all
also very active (Einhorn et al. 1988). These results make this combination of chemotherapy a standard
were subsequently reconfirmed by Fukuoka et al. for fit patients undergoing thoracic radiation therapy.
(1991) who alternated cyclophosphamide/doxorubi- Since it was well documented that cisplatin/etoposide
cin/vincristin and cisplatin/etoposide which were had less cardiac and lung toxicity when compared
shown to be superior to either cyclophosphamide/ to cyclophosphamide/doxorubicin/vincristin, it was
doxorubicin/vincristin alone or cisplatin/etoposide preferentially used with thoracic radiation therapy,
alone (median survival: 16.8 vs. 12.4 vs. 11.7 months). providing 2-year survivals of [40% (Turrisi et al.
The regimen containing cisplatin/etoposide has 1999; Takada et al. 2002). Although majority of
become the standard therapy for patients with small- investigators continue to consider cisplatin/etopop-
cell lung cancer, especially when combined with side and thoracic radiation therapy as the mainstay
radiation therapy. In a Phase III study, the cisplatin/ of concurrent treatment today, carboplatin was
etoposide appeared superior to cyclophosphamide, sometimes used instead of cisplatin (Kosmidis et al.
epirubicin, and vincristine in 436 patients randomized 1994; Jeremic et al. 1997). It was used in combi-
to either cisplatin/etoposide or cisplatin/etoposide/ nation with etoposide (i.e., carboplatin/etoposide)
vincristin. The 5-year survival rates were between 5 due to a similar response and survival as cisplatin/
and 2% in the cisplatin/etoposide and cisplatin/eto- etoposide but with less kidney and ear toxicity than
poside/vincristin arms, respectively (p = 0.0004). In cisplatin/etoposide (Kosmidis et al. 1994; Jeremic
subgroup analysis done for 214 patients with limited- et al. 1997). Other drugs were also attempted to
disease, this benefit was even more pronounced incorporate into the treatment plan (Woo et al.
(5-year survival; 10 vs. 3%; p = 0.0001), while for 2000; Hanna et al. 2002).
patients having extensive disease this benefit In an attempt to provide sustained and prolonged
remained unreported (Sundstrøm et al. 2002). duration of response and control of existing symp-
The use of cisplatin/etoposide in this disease has toms, some advocated to treat patients for the duration
been additionally supported by a systematic review of their life. Of a number of randomized trials which
using 36 randomized trials which have tested single have investigated this issue, only one study demon-
agent either cisplatin or etoposide or both (doublet) strated a survival advantage for limited-disease small-
against regimens not containing these agents. The cell cancer patients (Maurer et al. 1980). This is in a
significant improvement with use of these drugs in sharp contrast to numerous studies showing either no
comparison with chemotherapy with neither was advantage at all (Woods and Levi, 1984; Cullen et al.
demonstrated (Mascaux et al. 2000). Furthermore, a 1986; Bleehen et al. 1989; Lebeau et al. 1992;
meta-analysis of 19 trials that investigated the effects Giaccone et al. 1993; Beith et al. 1996; Sculier et al.
of chemotherapy with or without cisplatin in more 1996) or even showing detrimental effects of contin-
than 400 patients. Patients receiving cisplatin had sur- uous chemotherapy (Byrne et al. 1989). The lack of
vival advantage of 4.4% at 1 year (Pujol et al. 2000). survival improvement and increased toxicity of pro-
However, a recent systematic review of 29 trials longed treatment, made this approach having no role
involving 5530 patients compared platinum-based in the treatment of limited-disease small-cell lung
494 B. Jeremić et al.

cancer patients nowadays. Additionally, some studies myelosuppression, and five patients (8%) died from
investigated the optimal number of induction che- toxicity.Taken all together, the data from literature do
motherapy courses. Here, no survival benefit was seen not support any intensification of chemotherapy as
for 8 cycles of cyclophosphamide/etoposide/vincris- worthwhile further investigation nowadays.
tine compared to 4 cycles, if there was an option of a Last decade also brought investigation of the place
second line chemotherapy (Spiro et al. 1989). This and the role of the third generation drugs. In a Japan
was indirectly confirmed as early as in 1996 by pre- Clinical Oncology Group, phase III study in extensive
liminary results of an intergroup 0096 study which disease small-cell lung cancer, only (Noda et al. 2002)
produced convincing results with only 4 cycles of irinotecan was combined with cisplatin and compared
cisplatin/etoposide and thoracic radiation therapy to cisplatin/etoposide, with irinotecan/cisplatin arm
(Johnson et al. 1996a, b). It is, therefore, that current achieving significant survival advantage (the median
standard chemotherapy protocol is four cycles of a survival time, 390 vs. 287 days; 1-year survival, 58 vs.
platinum-based regimen, although advocates of more 38%; p = 0.002). Overall response rates were also
chemotherapy courses (e.g., six) continue to try to significantly higher in the irinotecan/cisplatin arm
obscure existing highest level of evidence by intro- (83 vs. 63%). Although high-grade diarrhea was seen
duction of data other than that coming from prospective only in irinotecan/cisplatin arm, high-grade hemato-
randomized clinical trials using combined (mostly logical toxicity was seen more frequently in the cis-
concurrent) radiation therapy and chemotherapy. platin/etoposide arm. Since topotecan was initially
Another approach was to intensify the dose of shown to be effective in relapsed small-cell lung cancer
chemotherapy which was tested in randomized trials patients, it was then evaluated in maintenance therapy
including either doxorubicin or alkylating-based after initial cisplatin/etoposide in chemonaive exten-
chemotherapy in the 1970s and 1980s (Cohen et al. sive disease small-cell lung cancer patients and com-
1977; Mehta et al. 1982; Figueredo et al. 1985), or pared to no maintenance therapy. The addition of
cisplatin-based in the 1990s (Arriagada et al. 1993), topotecan led to improvement in progression-free
occasionally including granulocyte colony-stimulat- survival but with no impact on survival (8.7 vs.
ing factor support (Ardizzoni et al. 2002). Improved 9.0 months, p = 0.71) (Schiller et al. 2001). Of tax-
survival was noted in the dose-intensive arm in 3 anes, only paclitaxel was tested in a phase III study.
studies, with two trials showing significant improve- With chemotherapy alone two recently published
ment. This was, however, accompanied with more studies compared cisplatin/etoposide with cisplatin/
severe toxicity, which influenced that the dose etoposide/paclitaxel. Both Mavroudis et al. (2001) and
intensification did not become standard treatment Niell et al. (2002) found no difference in response rates,
approach. The issue of the increase of the dose median, and overall survival, but observed more
intensity is also attempted by decreasing the interval treatment-related deaths in the cisplatin/etoposide/
between the cycles of chemotherapy. Two trials paclitaxel regimen. Gatzmeier et al. (2000) showed no
demonstrated an improvement in survival (Steward difference in toxicity between paclitaxel, carboplatin,
et al. 1998; Thatcher et al. 2000) but again, unfortu- and etoposide vs. carboplatin, etoposide, and vincris-
nately, accompanied by increased toxicity. Most tine in limited-disease and extensive disease small-cell
recently, Leyvraz et al. (2008) randomly assigned lung cancer. Finally, a preliminary analysis of another
patients who had limited or extensive small-cell lung study (Birch et al. 2000) showed only modest
cancer with no more than two metastatic sites to high- improvements in the overall response rate with a trend
dose or standard-dose chemotherapy with ifosfamide, toward improvement in survival when paclitaxel was
carboplatin, and etoposide. High-dose chemotherapy added to carboplatin, and etoposide and compared to
cycles included bone marrow support. The primary carboplatin and etoposide in patients with extensive
outcome was 3-year survival. The 3-year survival rates disease small-cell lung cancer. Data from four phase II
were 18 and 19% in the high-dose and standard- trials in small-cell lung cancer showed only moderate
dose chemotherapy arms, respectively. No differences success when paclitaxel was added to concurrent
were observed between the two arms in overall cisplatin/etoposide and thoracic radiation therapy
response and complete response, respectively. High- (Levitan et al. 2000; Ettinger et al. 2005; Sandler et al.
dose treatment was predictably associated with severe 2000; Bremnes et al. 2001), with complete response
Limited-Disease Small-Cell Lung Cancer 495

rates of 13–81% and median survival times of about Of the remaining issues in thoracic radiation therapy,
22 months. Finally, recent analysis of the Southwest timing of combined radiation therapy and chemo-
Oncology group phase II study 9713 provided another therapy, and total dose and fractionation used, have
set of the data on the use of paclitaxel in 87 patients with attracted most of the attention of researchers. When
limited-disease small-cell lung cancer (Edelmen et al. timing of combined radiation therapy and chemo-
2004). Concurrent cisplatin/etoposide/radiation ther- therapy is considered, it is usually defined as either
apy part of the combined modality program was fol- concurrent, or sequential or alternating. While some
lowed by three cycles of consolidation paclitaxel/ of the initial studies showed promising results for
carboplatin. The response rate was 86%, the median alternating radiation therapy and chemotherapy, this
survival time was 17 months and a 2-year survival rate type of combined approach is mostly abandoned
was 33%, while the progression-free survival at 2 years today. The main question with the remaining two
was only 21%. This prompted authors to conclude that modes of administration is simply whether any por-
paclitaxel is inactive against small-cell lung cancer and tion of thoracic radiation therapy and chemotherapy
suggested its abandoning in further investigation, overlaps and, if this is the case, when overlapping
confirming previously disappointing results of the occurs. Early concurrent thoracic radiation therapy
Eastern Cooperative Oncology Group (Sandler et al. and chemotherapy studies used non-platinum regi-
2000). Contrary to these results, European Organiza- mens, or alternated it with cisplatin/etoposide, while
tion for research and treatment of Cancer (Reck et al. more recent ones were exclusively platinum-based
2003) found an advantage for paclitaxel-containing regimens. Some studies (Perry et al. 1987; Schultz
arm in patients with small-cell lung cancer. In limited- et al. 1988; Work et al. 1997) suggested that thoracic
disease small-cell lung cancer patients it achieved the radiation therapy delayed until the fourth cycle of
median survival time of 17.6 months, which was quite chemotherapy (Perry et al. 1987) or until day 120
similar to that of the Southwest Oncology group study (Schultz et al. 1988) may be superior to initial radi-
cited above as well as those achieved during the ation therapy or suggested no difference when com-
Intergroup study (Turrisi et al. 1999), and somewhat pared to early thoracic radiation therapy and
lower than achieved in other prospective randomized chemotherapy (Work et al. 1997). Likely the expla-
studies which used only cisplatin/etoposide combina- nation lies in marked reduction of chemotherapy dose
tion (Jeremic et al. 1997; Takada et al. 2002). in the Cancer and Leukemia Group B (Perry et al.
As a summary, there is no firm basis to recommend 1987) and the Danish trial when thoracic radiation
either dose intensification or the integration of new therapy was applied early. It is important to note that
drugs into actual regimens, due to the risk of severe the Danish trial (Work et al. 1997) can not really be
toxicity and the lack of clearly demonstrated considered as a concurrent study because sequential
improvement in overall survival. This is especially so radiation therapy was used before and after chemo-
when one considers lack of the data for chemotherapy therapy. More recent studies using cisplatin/etoposide
combined with thoracic radiation therapy. Targeted (Jeremic et al. 1997; Takada et al. 2002) or cisplatin/
agents are now widely under testing in this disease in etoposide alternating with cyclophosphamide/doxo-
chemotherapy-only setting. If proven beneficial, it is rubicin/vincristin (Murray et al. 1993) showed clear
reasonable to expect their testing within the concur- superiority for early administration of thoracic radi-
rent radiation therapy-chemotherapy approach. ation therapy (concurrently given during the first or
the second cycle of chemotherapy). Overview of
existing trials is presented in Table 1. These studies
have also reconfirmed in clinical practice an original
4 Thoracic Radiation Therapy Goldie and Coldman (1979) theoretical consider-
ations that an early administration of both treatment
Among several important issues investigated in recent modalities lead to the best outcome on both local and
decades, prophylactic cranial irradiation seems to distant level (Table 1). Early concurrent thoracic
have been settled. It will be a matter of another radiation therapy and cisplatin/etoposide chemother-
chapter of this book. In addition, treatment volumes apy was capable of achieving 5-year survival of
will also be a subject of another chapter of this book. [20%, while late thoracic radiation therapy usually
496 B. Jeremić et al.

Table 1 Randomized phase III trials investigating optimal timing of combined thoracic radiation therapy and chemotherapy in
limited-disease small-cell lung cancer
Author Year N CHT RT RT timing MST Survival Outcome
(weeks) (months) (5 year) (%) (p)
Perry 1987 125 CEVA 50 Gy/24 fr/ 1 13.04 7
et al. QD
145 same Same 9 14.54 13 0.08
Murray 1993 155 CAV/PE 40 Gy/15 fr/ 3 21.2 20
et al. QD
153 same 15 16.0 11 0.008
Work 1997 99 PE ? CAV 40–45 Gy/22 1 10.7 11
et al. fr/QD
100 same same 18 12.9 12 0.4
Jeremic 1997 52 PE (CpE with 54 Gy/36 fr/ 1 34 30
et al. RT) BID
51 same same 6 26 15 0.052
Takada 2002 114 PE 45 Gy/30 fr/ 1 27.2 24
et al. BID
114 same same 15 19.7 18 0.097
Spiro 2006 159 CAV/PE 40 Gy/15fx/ 3 13.7 16*
et al. QD
166 same same 15 15.1 22* 0.23
CEV cyclophosphamide, etoposide, vincrstine; CAV cyclophosphamide, doxorubicin, vincristine; PE cisplatin, etoposide; CpE
carboplatin, etoposide; fr fraction; RT radiation therapy;* at 3 years

Table 2 Timing/scheduling of RT and CHT in LD SCLC: systematic reviews/meta-analyses


Author Year Time (year) OS RR 95% CI p
Fried et al. 2004 2 1.17 1.021.35 0.03
3 1.13 0.92–1.39 0.2
Huncharek/McGarry 2004 2 1.60 1.29–1.99 \0.01
3 1.49 1.15–1.93 \0.01
Pijls-Johannesma et al. 2005 2–3 0.84 0.56–1.28 0.4
5 0.80 0.47–1.38 0.4
OS overall survival, RR risk ratio, CI confidence interval

obtained only about 10%. Therefore, it became a Recently, several meta-analyses and systematic
common practice to offer as early as possible (cycle reviews addressed this issue by putting all existing
one or two of chemotherapy) thoracic radiation ther- evidence into a perspective of timing of combined
apy with curative doses worldwide. Others have also radiation therapy and chemotherapy in limited-dis-
proved that this is indeed the fact even outside the ease small-cell lung cancer (Table 2). (Huncharek
clinical trial, e.g., in an institutional setting. Kamath and McGarry 2004) observed significantly superior
et al. (1998) showed in a small study on 48 patients survival at both 2- and 3-years for early radiation
that early concurrent thoracic radiation therapy/cis- therapy and chemotherapy. Fried et al. (2004)
platin/etoposide offers advantage over sequential observed a significantly higher 2-year survival in
chemotherapy and thoracic radiation therapy in terms the early group and there was a suggestion of a
of overall survival and decreased distant metastasis in similar trend at 3 and 5 years. Contrary to these,
patients with limited-disease small-cell lung cancer. Pijls-Johannesma et al. (2005) did not find any
Limited-Disease Small-Cell Lung Cancer 497

advantage for early radiation therapy and chemo- Regarding thoracic radiation therapy dose and
therapy. Finally, Spiro et al. (2006) found no differ- fractionation, total doses used for limited-disease
ence between the early and the late administration of small-cell lung cancer were usually about 50 Gy,
the two regimens. However, they have disclosed that standard fractionation. They have, however, been as
test for heterogeneity was significant (p = 0.0002), low as 30 Gy and as high as 70 Gy. In addition, many
which indicated that hazard ratios estimates likely recent studies have used some form of hyperfrac-
differ from overall estimates. To correct these ambi- tionation (b.i.d.). Whichever fractionation regimen
guities, they have performed Forrest plot analysis for one uses, even in the era of concurrent thoracic
treatment effect. When they focused on cases which radiation therapy and chemotherapy, one major site of
received all chemotherapy cycles, they have found recurrence continues to be in-field (about 30% are
better survival for patients in early group if patients isolated and additional 20% are combined with sys-
received similar percentage of chemotherapy in both temic progression). Majority of available studies are
arms, contrary to cases when there was less percent- retrospective in nature, with one study (Choi and
age in early arm, leading to better survival in late Carry 1989) observing a better local control for doses
group. Similarly, positive effect of hyperfractionated 40–50 Gy than with doses \40 Gy ([50 vs. 30%),
radiation therapy was found in early group, but not in indicating, therefore, potential dose–response rela-
late, as well as when platinum based chemotherapy tionship. Another study indicated excellent local
was used, early group was better and when not, late control after 60 Gy, being 97% (Papac et al. 1987).
was better. These 4 analyses (Huncharek and The Cancer and Leukemia Group B (Choi et al. 1998)
McGarry 2004; Fried et al. 2004; Pijls-Johannesma performed a trial to identify at least 70 Gy (using
et al. 2005; Spiro et al. 2006) brought somewhat standard fractionation) as the maximum tolerated
conflicting results which were result of: (a) different dose for combination with chemotherapy. Subse-
definition of limited-disease small-cell lung cancer, quently, they (Bogart et al. 2004) reported that 70 Gy
(b) different definition of ‘‘early’’ and ‘‘late’’ admin- was feasible and effective when given concurrently
istration, (c) inclusion of ‘‘grey literature’’, (d) dif- with 3 cycles of carboplatin and etoposide, following
ferent patient number, and (e) lack of individual an induction with 2 cycles of paclitaxel and topotec-
patient data. In an attempt to resolve the matter, an, with the median overall survival of 19.8 months
Jeremic (2006) performed ‘‘meta analysis’’ of the with a 1-year survival rate of 70% the median failure-
meta-analyses, identifying common findings in free survival was 12.9 months. Group study results
existing analyses. The finding included the following: were also confirmed in a single-institutional setting.
(a) there was more leucopoenia in late group, (b) there Miller et al. (2003) retrospectively evaluated the data
was a favorable effect of short (B30 days) overall of 65 patients from the Duke University in which
treatment time, (c) there was a favorable effect of 58–66 Gy, standard fractionation was used with either
hyperfractionation, (d) there was a favorable effect of concurrent (n = 32) or sequential (n = 33) chemo-
platinum-etoposide chemotherapy, and (e) there was therapy. Somewhat lower (30%) 2-year survival rate
negative effect of split-course radiation therapy. was explained by less than one-half of patients
Overall, prevailing evidence is that nowadays, using receiving concurrent thoracic radiation therapy and
‘‘standard’’ approach consisting of hyperfractionated chemotherapy and only 26% received prophylactic
radiation therapy and 4 courses of chemotherapy cranial irradiation. The toxicity of their regimen was
based on cisplatin-etoposide, early administration low. Similarly, Roof et al. (2003) observed that
seems favorable and should be practiced as standard overall survival, local control, and disease-free sur-
approach. Reports showing that prolonged (e.g., 4–6 vival obtained with [50 Gy compared favorably with
cycles) sequential administration of chemotherapy the historic controls which were using lower doses.
followed by radical thoracic radiation therapy is Most recently, Komaki et al. (2003) reported on
inferior treatment approach when compared to early Radiation Therapy Oncology Group 9712 study
and concurrent radiochemotherapy is unfortunately which was a phase I dose-escalation study of thoracic
still occurring nowadays (El-Sharouni et al. 2009; radiation therapy with concurrent cisplatin/etoposide
Yilmaz et al. 2010). in limited-disease small-cell lung cancer. Thoracic
498 B. Jeremić et al.

radiation therapy was given 1.8 Gy daily to 36 Gy fractionation, providing total dose and treatment
followed by boost delivered with escalations of duration being similar if not the same). Extending
1.8 Gy b.i.d. during the final days which permitted overall treatment time, therefore, which allows tumor
doses of up to 64.8 Gy to be given. The maximum cell regeneration, may have been the reason for this
tolerated dose was determined to be 61.2 Gy in 34 finding due to a delay in thoracic radiation therapy
fractions of 1.8 Gy when given concurrently with 2 either by long lasting induction chemotherapy or by
cycles of cisplatin/etoposide and followed by 2 split-course protocol for thoracic radiation therapy. A
additional cycles of cisplatin/etoposide. quality-adjusted reanalysis of a that phase III trial
Besides hyperfractionation and conventional frac- (Bonner et al. 1999; Schild et al. 2003) comparing
tionation, hypofractionated radiation therapy regi- once-daily thoracic radiation versus twice-daily
mens were also used, which was thought to cause thoracic radiation in patients with limited-stage small-
more damage to small-cell lung cancer cells (Murray cell lung cancer using Quality Time Without Symp-
et al. 1993; Spiro et al. 2006). Interestingly, shifting toms or Toxicity methodology showed no difference
from such hypofractionated to conventionally frac- in survival after adjusting for toxicity and progression
tionated thoracic radiotherapy did not alter outcomes, (Sloan et al. 2002).
the survival, local control, and toxicity rates were all While accelerated hyperfractionated thoracic
similar (Videtic et al. 2003). Of altered fractionated radiation therapy was practiced with increasing evi-
regimens, accelerated hyperfractionation was the dence in the last two decades, the accumulated data
logical choice due to a high sensitivity of small-cell show different outcome (Johnson et al. 1996a, b; Ali
lung cancer to radiation therapy, sparing effect of et al. 1998; Mennecier et al. 2000; Segawa et al.
twice-daily fractionation and possible effect of the 2003) and toxicity profile. The future studies directly
dose acceleration to combat rapid proliferation comparing b.i.d. to once-daily fractionation will bring
thought to occur in small-cell lung cancer. In the definitive answers about optimal total dose and frac-
Intergroup study (Johnson et al. 1996a, b; Turrisi et al. tionation regimen preferentially used. Currently, two
1999), 45 Gy given in 30 fractions in 3 weeks major clinical trials investigating this issue are
(1.5 Gy b.i.d. fractionation) was compared with the recruiting patients. In a CONVERT trial, EORTC is
same dose given once-daily, both with concurrent evaluating 66 Gy using standard fractionation with
cisplatin-etoposide chemotherapy. While a survival the bid fractionation as used in the Intergroup study
was significantly better in the b.i.d. arm (5-year, 26 (45 Gy in 30 fractions in 15 treatment days in
vs. 19%), this was, however, achieved with somewhat 3 weeks). Similarly, joint CALGB 30610/RTOG
higher incidence of acute toxicity. Another study 0538 is directly comparing the same control Inter-
investigating this issue was a North Central Cancer group regimen with two experimental arms, either
Treatment Group study which compared concurrent conventional (QD) or concomitant boost regimen
two cycles of cisplatin/etoposide with either b.i.d., (CB). The better of the two experimental arms (CB) is
split-course thoracic radiation therapy (48 Gy in a then directly compared to hyperfractionated regimen.
total of 5.5 weeks) or once-daily thoracic radiation Mature data from the two trials should supplement
therapy (50.4 Gy), both given after 3 cycles of cis- existing ones and hopefully give better perspective
platin/etoposide (Bonner et al. 1999). There was no about fractionation issue. Recent single-institutional
difference in a 3-year overall and locoregional con- report (Watkins et al. 2010a) compared two radiation
trol. After 5 years (Schild et al. 2003), the median and therapy regimens with planned doses of (1) [59.4 Gy
5-year survival were 20.4 months and 22% for b.i.d. at 1.8–2.0 Gy per once-daily fraction or (2) [45 Gy
versus 20.5 months and 21% for once-daily thoracic at 1.5 Gy b.i.d. with concurrent platinum-based che-
radiation therapy, respectively (p = 0.7). Having motherapy. A total of 71 patients were included in the
these two studies together, possible explanation may study with patient, tumor, staging, and treatment
lie either in inferiority of split-course regimen (which factors being similar between the two treatment
undermined the effect of hyperfractionation) or effects groups. Acute toxicities were similar between the
of acceleration outweighing those of hyperfractiona- groups. The 2-year overall survival estimates were
tion (To put it in other words, hyperfractionation similar at 43 and 49% for the once-daily versus twice-
given with a split equals conventional, once-daily daily groups, respectively. Isolated in-field failures
Limited-Disease Small-Cell Lung Cancer 499

were similar between the two groups. While this Mira and Livingston (1980) found that the majority
analysis did not detect a statistically significant who failed in the chest also failed at outside the field
difference in acute toxicities, disease control, or sur- (lung periphery) but not within nodal structures. It
vival outcomes in limited-stage small-cell lung cancer suggested that prechemotherapy volumes would pro-
patients treated with concurrent chemotherapy and vide improved local control and, therefore, be pref-
once-daily versus twice-daily radiation therapy, it erentially used in this setting. In contrast to it, in the
should not be forgotten that other regimens of b.i.d. study of Liengswangwong et al. (1994) the use of
irradiation (e.g., 54 Gy in 36 fractions in 18 treatment postchemotherapy volumes was supported. Of a total
days in 3.5 weeks) have been successfully imple- of 59 patients studied, 28 were treated with postche-
mented in practice concurrently with low-dose che- motherapy tumor volumes and 31 with prechemo-
motherapy in both limited-disease (Jeremic et al. therapy tumor volumes. Of a total group, 10 of 31
1997) and extensive disease small-cell lung cancer patients treated with radiation therapy fields that
(Jeremic et al. 1999), providing not only excellent encompassed pre- chemotherapy tumor volumes and
results, but also leading to low toxicity. Finally, it is 9 of 28 patients treated with radiation therapy fields
not unreasonable to expect that novel drugs eagerly that encompassed postchemotherapy tumor volumes
await its place and time in this disease and the data had locoregional failures, suggesting no difference for
slowly emerge (Sandler et al. 2000). choice of irradiation volume. These results indicate
There are two important issues one must take into that in cases of bulky disease within pulmonary
account when considering the irradiation volume for parenchyma shrinking after chemotherapy one may
limited-disease small-cell lung cancer patients. The use radiation therapy fields to encompass postche-
first is related to the irradiation of the pre- or post- motherapy findings in order to reduce the treatment
chemotherapy disease volume and the second one is toxicity, especially in frail patients and/or those with
related to the elective nodal irradiation. When radia- limited pulmonary reserve. None of the landmark
tion therapy starts concurrently with the first cycle of trials on thoracic radiotherapy in limited-disease
chemotherapy this issue simply does not exists and small cell-lung cancer provide a consistent basis for
one treats what available imaging says. The question drawing conclusions on the role of elective nodal
of including or not (in the radiation therapy field) irradiation in small-cell lung cancer. Some were using
regions where the visible tumor involvement was elective nodal irradiation (Jeremic et al. 1997; Takada
presumably sterilized with chemotherapy arises as a et al. 2002; Turrisi et al. 1999), while some were not
problem in cases of delayed radiation therapy, when it (Murray et al. 1993; Work et al. 1997; Spiro et al.
is given concurrently with 2 or 3 cycles of chemo- 2006). Ongoing studies, the CONVERT and Inter-
therapy. Only one randomized clinical trial has group randomized Phase III trials, are addressing a
examined the issue of radiotherapy treatment volume question of dose but not the use of elective nodal
in small-cell lung cancer. This study, performed by irradiation. To date, we have only one prospective
southwest oncology group (Kies et al. 1987), involved Phase II study directly addressing the issue of elective
466 patients and randomized patients with a partial nodal irradiation in limited-disease small-cell lung
response or stable disease after four cycles of non- cancer (de Ruysscher et al. 2006). The authors pro-
platinum-based chemotherapy to radiation therapy spectively evaluated the patterns of recurrence when
fields based either on the pre- or on post-chemother- elective nodal irradiation was not used. In total, 27
apy volume of disease. No statistical differences in patients received hyperfractionated thoracic radiation
survival or recurrence patterns were noted as a func- therapy of 45 Gy in 30 fractions concurrent with
tion of volume treated. Postchemotherapy volumes carboplatin and etoposide. Only the primary tumor
were, therefore, judged appropriate for target delin- and the positive lymph nodes on the pretreatment CT
eation, not risking higher incidence of marginal fail- scan were irradiated. After a median follow-up of
ures. Several retrospective analyses have assessed 18 months, seven patients developed a local recur-
pre- versus post-chemotherapy tumor volumes with rence. Three patients developed an isolated nodal
contrasting results (Perez et al. 1981; White et al. failure, all of them in the ipsilateral supraclavicular
1982). In 17 limited-disease small-cell lung cancer fosse. The authors concluded that omission of elective
patients treated with postchemotherapy volumes, nodal irradiation on the basis of CT scans in patients
500 B. Jeremić et al.

with limited-disease small-cell lung cancer resulted in because they resembled limited-disease small-cell
a higher rate than expected rate of isolated nodal lung cancer patients at most. In them, after 3 initial
failures in the ipsilateral supraclavicular fosse and the cycles of cisplatin/etoposide, accelerated hyperfrac-
intentional omission of elective nodal irradiation may tionated thoracic radiation therapy offered survival
not be safe and should not be practiced outside the advantage over that achieved with chemotherapy
clinical trials. Contrary to that, recent retrospective alone (the median survival time : 17 vs. 11 months;
study of Watkins et al. (2010b) showed that involved- 5 year survival rates : 9.1 vs. 3.7%, respectively;
field thoracic radiation therapy given with concurrent p = 0.041) sue to an improvement in the local
chemotherapy did not appear to have an adverse recurrence-free survival (p = 0.062). Patients treated
impact on the anticipated patterns of failure, disease with thoracic radiation therapy achieved better results
control, or overall survival in patients with limited- than those treated with chemotherapy only regarding
disease small-cell lung cancer. In the most compre- both median time to first relapse (13 vs. 9 months,
hensive review of the use of elective nodal irradiation respectively) and 1–5 year first relapse-free survival
in limited-disease small-cell lung cancer, authors (p = 0.045). Interestingly, after initial 3 cycles of
recently noted the absence of strong evidence sup- cisplatin/etoposide, thoracic radiation therapy offered
porting omission of elective nodal irradiation. They higher response rate than additional cisplatin/etopo-
have suggested that clinicians must carefully balance side. When further response was evaluated, additional
the increased failure risk, expected to occur with cisplatin/etoposide (in both groups) offered nothing
omission of elective nodal irradiation with the reduc- but a few percent of additional response, an indirect
tion of treatment-related toxicities with involved fields evidence of the necessity of limiting of the number of
in the decision-making process (Videtic et al. 2008). chemotherapy cycles to 4–6. Results of this study
await further verification, an important task for the
future endevors in small-cell lung cancer.
5 Conclusions

The standard treatment for the majority of patients


with limited-disease small-cell lung cancer is com- References
bination of thoracic radiation therapy and cisplatin/
etoposide, given concurrently, with thoracic radiation Ali MA, Kraut MJ, Valdivieso M, Herskovic AM, Du W,
Kalemkerian GP (1998) Phase II study of hyperfractionated
therapy being started early. While majority of insti-
radiotherapy and concurrent weekly alternating chemother-
tutions worldwide use 4 cycles of cisplatin/etoposide, apy in limited-stage small cell lung cancer. Lung Cancer
numerous thoracic radiation therapy and chemother- 22:39–44
apy issues remain unsolved. Ongoing studies will help Amarasena IU, Walters JA, Wood-Baker R, Fong K (2008)
clear these important issues in optimizing the treat- Platinum versus non-platinum chemotherapy regimens for
small cell lung cancer. Cochrane Database Syst
ment approach and outcome in this disease. The les- Rev.4:CD006849
sons we have learned from optimization of the Ardizzoni A, Tjan-Heijnen VCG, Postmus PE, Buchholz E,
treatment approach in limited-disease small-cell lung Biesma B, Karnicka-Mladowska H, Dziadziuszko R,
cancer also served as an attempt to optimize the Burghouts J, van Meerbeck JP, Gans S, Legrand C,
Debruyne C, Giaccone G, Manegold C (2002) Standard
treatment in extensive disease small-cell lung cancer. versus intensified chemotherapy with granulocyte colony-
As we have recently shown in a prospective ran- stimulating factor support in small-cell lung cancer: a
domized trial, thoracic radiation therapy can play an prospective European Organization for research and treat-
important role in extensive disease small-cell lung ment of cancer—lung cancer group phase III trial–08923.
J Clin Oncol 20:3947–3955
cancer, providing that suitable patients are identified Arriagada R, Le Chevalier T, Pignon JP, Riviere A, Monnet I,
(Jeremic et al. 1999). We have focused on those Chomy P, Tuchais C, Tarayre M, Ruffie P (1993) Initial
patients who have the most favorable prognosis after chemotherapeutic doses and survival in patients with
induction chemotherapy, i.e., those achieving com- limited small-cell lung cancer. N Engl J Med 25:1848–1852
Auperin A, Arriagada R, Pignon JP, Le Pechoux C, Gregor A,
plete response at distant sites accompanied with either Stephens RJ, Kristjansen PE, Johnson BE, Ueoka H,
complete response or partial response intrathoraci- Wagner H, Aisner J (1999) Prophylactic cranial irradiation
cally. They were chosen as a subject of our study for patients with small-cell lung cancer in complete
Limited-Disease Small-Cell Lung Cancer 501

remission prophylactic cranial irradiation overview collab- De Ruysscher D, Bremer RH, Koppe F, Wanders S, van Haren E,
orative group. N Engl J Med 341:476–484 Hochstenbag M, Geeraedts W, Pitz C, Simons J, ten Velde G,
Beith JM, Clarke SJ, Woods RL, Bell DR, Levi JA (1996) Dohmen J, Snoep G, Boersma L, Verschueren T, van
Long-term follow-up of a randomised trial of combined Baardwijk A, Dehing C, Pijls M, Minken A, Lambin P
chemoradiotherapy induction treatment, with and without (2006) Omission of elective node irradiation on basis of CT-
maintenance chemotherapy in patients with small cell scans in patients with limited disease small cell lung cancer: a
carcinoma of the lung. Eur J Cancer 32A:438–443 phase II trial. Radiother Oncol 80:307–312
Birch R, Greco F, Hainsworth J (2000) Preliminary results of a Edelmen MJ, Chansky K, Gaspar LE, Leigh B, Weiss GR,
randomized study comparing etoposide and carboplatin with Taylor SA, Crowley J, Livingston R, Gandara DR (2004)
or without paclitaxel in newly diagnosed small cell lung Phase II trial of cisplatin/etoposide and concurrent radio-
cancer. Proc Am Soc Clin Oncol 19:490 (Abstract) therapy followed by paclitaxel/carboplatin consolidation for
Bleehen NM, Fayers PM, Girling DJ, Stephens RJ (1989) limited small-cell lung cancer: southwest Oncology Group
Controlled trial of twelve versus six courses of chemother- 9713. J Clin Oncol 22:127–132
apy in the treatment of small-cell lung cancer. Br J Cancer Einhorn LH, Crawford J, Birch R, Omura G, Johnson DH,
59:584–590 Greco FA (1988) Cisplatin plus etoposid consolidation
Bogart JA, Herndon JE, Lyss AP, Watson D, Miller AA, Lee following cyclophosphamide, doxorubicin, and vincristine
ME, Turrisi AT, Green MR, Cancer and Leukemia Group B in limited small-cell lung cancer. J Clin Oncol 6:451–456
study 39808 (2004) 70 Gy thoracic radiotherapy is feasible El Sharouni SY, Kal HB, Barten-Van Rijbroek A, Struikmans H,
concurrent with chemotherapy for limited-stage small-cell Battermann JJ, Schramel FM (2009) Concurrent versus
lung cancer: analysis of cancer and leukemia group B study sequential chemotherapy and radiotherapy in limited disease
39808. Int J Radiat Oncol Biol Phys 59:460–468 small cell lung cancer: a retrospective comparative study.
Bonner JA, Sloan JA, Shanahan TG, Brooks BJ, Marks RS, Anticancer Res 29:5219–5224
Krook JE, Gerstner JB, Maksymiuk A, Levitt R, Mailliard JA, Ettinger DS, Berkey BA, Abrams RA, Fontanesi J, Machtay M,
Tazelaar HD, Hillman S, Jett JR (1999) Phase III comparison Duncan PJ, Curran WJ Jr, Movsas B, Byhardt RW,
of twice-daily split-course irradiation versus once-daily Radiation Therapy Oncology Group 9609 (2005) Study of
irradiation for patients with limited stage small-cell lung paclitaxel, etoposide, and cisplatin chemotherapy combined
carcinoma. J Clin Oncol 17:2681–2691 with twice-daily thoracic radiotherapy for patients with
Bremnes RM, Sundstrom S, Vilsik J, Aasebo U (2001) limited-stage small-cell lung cancer: a radiation therapy
Multicenter phase II trial of palcitaxel, cisplatin, and oncology group 9609 phase II study. J Clin Oncol
etposide with concurrent radiation for limited-stage small 23:4991–4998
cell lung cancer. J Clin Oncol 19:3532–3538 Figueredo AT, Hryniuk WM, Strautmanis I, Frank G, Rendell S
Byrne MJ, Van Hazel G, Trotter J, Cameron F, Shepherd J, (1985) Cotrimoxazole prophylaxis during high-dose che-
Cassidy B, Gebski V (1989) Maintenance chemotherapy in motherapy of small-cell lung cancer. J Clin Oncol 3:54–64
limited small cell lung cancer: a randomised controlled Fried DB, Morris DE, Poole C, Rosenman JG, Halle JS,
clinical trial. Br J Cancer 59:584–590 Detterbeck FC, Hensing TA, Socinski MA (2004) System-
Choi NC, Carey RR (1989) Importance of radiation dose in atic review evaluating the timing of thoracic radiation
achieving improved locoregional tumor control in small-cell therapy in combined modality therapy for limited-stage
lung carcinoma: an update. Int J Radiat Oncol Biol Phys small-cell lung cancer. J Clin Oncol 22:4837–4845
17:307–310 Fukuoka M, Furuse K, Saijo N, Nishiwaki Y, Ikegami H,
Choi N, Herndon J, Rosenman J, Carey RW, Chung CT, Tamura T, Shimoyama M, Suemasu K (1991) Randmoized
Bernard S, Leone L, Seagren S, Green M (1998) Phase I trial of cyclophosphamide, doxorubicin, and vincristine
study to determine the maximum tolerated dose of radiation versus cisplatin and etoposide versus alternation of these
in standard daily and accelerated twice daily radiotherapy regimens in small-cell lung cancer. J Natl Cancer Inst
schedules with concurrent chemotherapy for limited stage 83:855–861
small cell lung cancer : CALGB 8837. J Clin Oncol Gatzmeier U, von Pawel J, Macha H (2000) A phase III trial of
16:3528–3536 taxol, etoposide phosphate and carboplatin (TEC) versus
Cohen MH, Broder LE, Fossieck BE, Ihde DC, Minna JD carboplatin, etoposide phosphate, and vincristive (CEV) in
(1977) Intensive chemotherapy of small cell bronchogenic previously untreated small cell lung cancer. Proc Am Soc
carcinoma. Cancer Treat Rep 61:349–354 Clin Oncol 19:483 (Abstract)
Cohen MH, Ihde DC, Bunn PA Jr, Fossieck BE Jr, Matthews MJ, Giaccone G, Dalesio O, McVie GJ, Kirkpatrick A, Postmus PE,
Shackney SE, Johnston-Early A, Makuch R, Minna JD Burghouts JT, Bakker W, Koolen MG, Vendrik CP,
(1979) Cyclic alternating combination chemotherapy for Roozendaal KJ (1993) Maintenance chemotherapy in
small cell bronchogenic carcinoma. Cancer Treat Rep small-cell lung cancer: long-term results of a randomized
62:163–170 trial. J Clin Oncol 11:1230–1240
Cullen M, Morgan D, Gregory W, Robinson M, Cox D, Goldie JG, Coldman AJ (1979) A mathematical model for
McGivern D, Ward M, Richards M, Stableforth D, relating the drug sensitivity of tumors to their spontaneous
Macfarlane A (1986) Maintenance chemotherapy for ana- mutation rate. Cancer Treat Rep 63:1727–1735
plastic small cell carcinoma of the bronchus: a randomised, Green RA, Humphrey E, Close H, Patno ME (1969) Alkylating
controlled trial. Cancer Chemother Pharmacol 17:157–160 agents in bronchogenic carcinoma. Am J Med 46:516–525
Curran WJ (2001) Combined-modality therapy for limited- Greenlee RT, Murray T, Bolden S, Wingo PA (2000) Cancer
stage small cell lung cancer. Sem Oncol 28:14–22 statistics 2000. CA Cancer J Clin 50:7–33
502 B. Jeremić et al.

Hanna N, Ansari R, Fisher W, Shen J, Jung S-H, Sandler A Lebeau B, Chastang CL, Allard P, Migueres J, Boita F, Fichet D
(2002) Etoposide, ifosfamide and cisplatin (VIP) plus (1992) Six vs twelve cycles for complete responders to
concurrent radiation therapy for previosuly untreated lim- chemotherapy in small cell lung cancer: definitive results of
ited small cell lung cancer (SCLC) : a Hoosier oncology a randomised clinical trial. Eur Respir J 5:286–290
group (HOG) phase II study. Lung Cancer 35:293–297 Levitan N, Dowlati A, Shina D, Craffey M, Mackay W, DeVore R,
Huncharek M, McGarry R (2004) A meta-analysis of the timing Jett J, Remick SC, Chang A, Johnson D (2000) Multi-
of chest irradiation in the combined modality treatment of institutional pahse I/II trial of paclitaxel, cisplatin, and
limited-stage small cell lung cancer. Oncologist 9:665–762 etoposide with concurrent radiation for limited-stage small-
Ihde DC, Pass HI, Glatstein EJ (1993) Small cell lung cancer. cell lung carcinoma. J Clin Oncol 18:1102–1109
In: DeVita V, Hellman S, Rosenberg S (eds) Cancer— Leyvraz S, Pampallona S, Martinelli G, Ploner F, Perey L,
principles and practice of oncology. JB Lipincott Co., Aversa S, Peters S, Brunsvig P, Montes A, Lange A, Yilmaz
Philadelphia, pp 723–758 U, Rosti G; Solid Tumors Working Party of the European
Jeremic B (2006) Timing of concurrent radiotherapy and Group for Blood and Marrow Transplantation (2008) A
chemotherapy in limited-disease small-cell lung cancer: threefold dose intensity treatment with ifosfamide, carbo-
‘‘meta-analysis of meta-analyses’’. Int J Radiat Oncol Biol platin, and etoposide for patients with small cell lung
Phys 64:981–982 cancer: a randomized trial. J Natl Cancer Inst 100:533–541
Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1997) Mascaux C, Paesmans M, Berghmans T, Branle F, Lafitte JJ,
Initial versus delayed accelerated hyperfractionated radia- Lemaitre F, Meert AP, Vermylen P, Sculier JP, European
tion therapy and concurrent chemotherapy in limited small Lung Cancer Working Party (ELCWP) (2000) A systematic
cell lung cancer. J Clin Oncol 15:893–900 review of the role of etoposide and cisplatin in the
Jeremic B, Shibamoto Y, Nikolic N, Milicic B, Milisavljevic S, chemotherapy of small cell lung cancer with methodology
Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999) assessment and meta-analysis. Lung Cancer 30:23–36
The role of radiation therapy in the combined modality Maurer LH, Tulloh M, Weiss R, Blom J, Leone L, Glidewell O,
treatment of patients with extensive disease small-cell lung Pajak TF (1980) A randomized combined modality trial on
cancer (ED SCLC): a randomized study. J Clin Oncol small cell carcinoma of the lung. Cancer 45:30–39
17:2092–2099 Mavroudis D, Papadakis E, Veslemes M, Tsiafaki X, Stavraka-
Johnson BE, Bridges JD, Sobezeck M, Gray J, Linnoila RI, kis J, Kouroussis C, Kakolyris S, Bania E, Jordanoglou J,
Gazdar AF, Hankins L, Steinberg SM, Edison M, Frame JN, Agelidou M, Vlachonicolis J, Georgoulias V (2001) A
Pass H, Nesbitt J, Holden D, Mulshine JL, Glatstein E, Ihde multicenter randomized clinical trial comparing paclitaxel-
DC (1996a) Patients with limited-stage small-cell lung cisplatin-etoposide versus cisplatin-etoposide as first-line
cancer treated with concurrent twice-daily chest radiother- treatment in patients with small-cell lung cancer. Ann Oncol
apy and etoposide/cisplatin followed by cyclophosphamide, 12:463–470
doxorubicin, and vincristine. J Clin Oncol 14:806–813 Mehta C, Vogl SE, Farber S (1982) High-dose cyclophospha-
Johnson DH, Kim K, Sause W (1996b) Cispaltin (p), etoposide mide (c) in the induction (ind) chemotherapy (ct) of small
(e) ? thoracic radiotherapy (TRT) administered once or cell lung cancer (sclc)–Minor improvements in rate of
twice daily (bid) in limited stage (LS) small cell lung caner remission and survival. Proc Am Assoc Cancer Res 23:165
(sclc): final report of intergroup trial 0096. Proc Am Soc Mennecier B, Jacoulet P, Dubiez A, Westeel V, Bosset JF,
Clin Oncol 15:374 Magnin V, Depierre A (2000) Concurrent cisplatin/etopo-
Kamath SS, McCarley DL, Zlotecki RA (1998) Decreased side chemotherapy plus twice daily thoracic radiotherapy in
metastasis and improved survival with early thoracic limited stage small cell lung cancer: a phase II study. Lung
radiotherapy and prophylactic cranial irradiation in com- Cancer 27:137–143
bined modality treatment of limited-stage small cell lung Miller KL, Marks LB, Sibley GS, Clough RW, Garst JL,
cancer. Radiat Oncol Investig 6:226–232 Crawford J, Shafman TD (2003) Routine use of approxi-
Kies MS, Mira JG, Crowley JJ, Chen TT, Pazdur R, Grozea PN, mately 60 Gy once-daily thoracic irradiation for patients
Rivkin SE, Coltman CA Jr, Ward JH, Livingston RB (1987) with limited-stage small-cell lung cancer. Int J Radiat Oncol
Multimodal therapy for limited small cell lung cancer: a Biol Phys 56:355–359
randomized study of indcution combination chemotherapy Mira JG, Livingston RB (1980) Evaluation and radiotherapy
with or without thoracic radiation in complete responders; implications of chest relapse patterns in small cell lung
and with widefield versus reduced voleume radiation in carcinoma treated with radiotherapy-chemotherapy: study
partial responders: a southwest oncology group study. J Clin of 34 cases and review of the literature. Cancer
Oncol 5:592–600 46:2557–2565
Komaki R, Swann S, Ettinger D (2003) Phase I dose-escalation Mulshine JL, Treston AM, Brown HP, Birer MJ, Shaw GL
study of thoracic irradiation with concurrent chemotherapy (1993) Initiators and promoters of lung cancer. Chest
for patients with limited small cell lung cancer (LSCLC): 103(Suppl. 1):4S–11S
radiation therapy oncology group (RTOG) protocol 9712. Murray N, Coy P, Pater J, Hodson I, Arnold A, Zee BC, Payne
Proc Am Soc Clin Oncol 21:462 (Abstract 2539) D, Kostashuk EC, Evans WK, Dixon P (1993) Importance
Kosmidis P, Samantas E, Fountzilas G, Pavlidis N, Aposto- of timing for thoracic irradiation in the combined modality
lopoulou F, Skarlos D (1994) Cisplatin/etoposide vs carbo- treatment of limited stage small cell lung cancer. J Clin
platin/etoposide and irradiation in small-cell lung cancer: a Oncol 11:336–344
randomized phase III study. Semin Oncol 21(Suppl. 6): Niell HB, Herndon JE, Miller AA (2002) Randomized phase III
23–30 intergroup trial (CALGB 9732) of etoposide and cisplatin
Limited-Disease Small-Cell Lung Cancer 503

with or without paclitaxel and G-CSF in patients with etoposide in extensive-stage small-cell lung cancer: E7593–
extensive stage small cell lung cancer. Proc Am Soc Clin a phase III trial of the eastern cooperative oncology group.
Oncol 21:293a J Clin Oncol 19:2114–2122
Noda K, Nishiwaki Y, Kawahara M, Negoro S, Sugiura T, Schultz HP, Nielsen OS, Sell A (1988) Timing of chest radiation
Yokoyama A, Fukuoka M, Mori K, Watanabe K, Tamura T, with respect to combination chemotherapy in small cell lung
Yamamoto S, Saijo N, The Japan Clinical Oncology Group cancer, limited disease. Lung Cancer 4:153 (Abstract)
(2002) Irinotecan plus cisplatin compared with etoposide Sculier JP, Paesmans M, Bureau G, Giner V, Lecomte J, Michel J,
plus cisplatin for extensive small-cell lung cancer. N Engl J Berchier MC, Van Cutsem O, Kustner U, Kroll F, Sergysels
Med 346:85–91 R, Mommen P, Klastersky J (1996) Randomized trial
Papac RJ, Son Y, Bien R, Tiedemann D, Keohane M, Yesner R comparing induction chemotherapy versus induction chemo-
(1987) Improved local control of thoracic disease in small- therapy followed by maintenance chemotherapy in small-cell
cell lung cancer with higher dose thoracic irradiation and lung cancer. J Clin Oncol 14:2337–2344
cyclic chemotherapy. Int J Radiat Oncol Biol Phys Segawa Y, Uoeka H, Kiura K, Tabata M, Takigawa N, Hiraki Y,
13:993–998 Watanabe Y, Yonei T, Moritaka T, Hiyama J, Hiraki S,
Perez CA, Krauss S, Bartolucci AA, Durant JR, Lowenbraun S, Tanimoto M, Harada M (2003) Phase I/II study of altered
Salter MM, Storaalsi J, Kellermeyer R, Comas F (1981) schedule of cisplatin and etoposide administration and
Thoracic and elective brain irradiation with concomitant or concurrent accelerated hyperfractionated thoracic radiother-
delayed multiagent chemotherapy in the treatment of apy for limited-stage small-cell lung cancer. Lung Cancer
localized small cell carcinoma of the lung: a randomized 41:13–20
prospective study by the southeastern cancer study group. Shepherd FA, Crowley J, van Houtte P, Postmus PE, Carney D,
Cancer 47:2407–2413 Chansky K, Shaikh Z, Goldstraw P (2007) The international
Perry MC, Eaton WL, Propert KJ, Ware JH, Zimmer B, association for the study of lung cancer lung cancer stagign
Chahinian AP, Skarin A, Carey RW, Kreisman H, Faulkner project proposals regarding teh clinical staging of small-cell
C (1987) Chemotherapy with or without radiation therapy in lung cancer in the forthcoming (seventh) edition of the
limited small-cell carcinoma of the lung. N Engl J Med tumor, node, metastasis classification for lung cancer.
316:912–918 J Thorac Oncol 2:1067–1077
Pignon JP, Arriagada R, Ihde DC, Johnson DH, Perry MC, Sloan JA, Bonner JA, Hillman SL, Allmer C, Shanahan TG,
Souhami RL, Brodin O, Joss RA, Kies MS, Lebeau B Brooks BJ, Marks RS, Vargas-Chanes D, Jett JR (2002) A
(1992) A meta-analysis of thoracic radiotherapy for small- quality-adjusted reanalysis of a phase III trial comparing
cell lung cancer. N Engl J Med 327:1618–1627 once-daily thoracic radiation vs twice-daily thoracic radia-
Pijls-Johannesma MC, De Ruysscher D, Lambin P, Rutten I, tion in patients with limited-stage small-cell lung cancer. Int
Vansteenkiste JF (2005) Early versus late chest radiotherapy J Radiat Oncol Biol Phys 52:371–381
for limited stage small cell lung cancer. Cochrane Database Spiro SG, Souhami RL, Geddes DM, Ash CM, Quinn H,
Syst Rev 1:CD004700 Harper PG, Tobias JS, Partridge M, Eraut D (1989)
Pujol JL, Carestia L, Daurès JP (2000) Is there a case for Duration of chemotherapy in small cell lung caner: a cancer
cisplatin in the treatment of small-cell lung cancer? A meta- research campaign trial. Br J Cancer 59:578–583
analysis of randomized trials of a cisplatin-containing Spiro SG, James LE, Rudd RM, Trask CW, Tobias JS, Snee M,
regimen versus a regimen without this alkylating agent. Gilligan D, Murray PA, Ruiz de Elvira MC, O’Donnell KM,
Br J Cancer 83:8–15 Gower NH, Harper PG, Hackshaw AK (2006) London lung
Reck M, von Pawel J, Macha HN (2003) Randomized phase III cancer group early compared with late radiotherapy in
trial of paclitaxel, etoposide, and carboplatin versus combined modality treatment for limited disease small-cell
carcoplatin, etoposide, and vincristine in patients with lung cancer: a London lung cancer group multicenter
small-cell lung cancer. J Natl Cancer Inst 95:1118–1127 randomized clinical trial and meta-analysis. J Clin Oncol
Roof KS, Fidias P, Lynch TJ, Ancukiewicz M, Choi NC (2003) 24:3823–3830
Radiation dose escalation in limited-stage small-cell lung Steward WP, von Pawel J, Gatzemaier U, Woll P, Thatcher N,
cancer. Int J Radiat Onmcol Biol Phys 57:701–708 Koschel G, Clancy L, Verweij J, de Wit R, Pfeifer W,
Sandler AB (2003) Chemotherapy for small cell lung cancer. Fennelly J, von Eiff M, Frisch J (1998) Effects of
Sem Oncol 30:9–25 granulocyte colony-stimulating factor and dose-intensifica-
Sandler A, Declerck L, Wagner H (2000) A phase II study of tion of V-ICE chemotherapy in small-cell lung cancer: a
cisplatin plus etoposide plus paclitaxel and concurrent prospective randomised study of 300 patients. J Clin Oncol
radiation therapy for previously untreated limited stage 16:642–650
small cell lung cancer (E2596): an eastern cooperative Sundstrøm S, Bremnes RM, Kaasa S, Aasebø U, Hatlevoll R,
oncology group trial. Proc Am Soc Clin Oncol 19: 491a Dahle R, Boye N, Wang M, Vigander T, Vilsvik J,
(Abstract 1920) Skovlund E, Hannisdal E, Aamdal S (2002) Norwegian
Schild S, Brindle JS, Geyer SM (2003) Long-term results of a lung cancer study group cisplatin and etoposide regimen is
phase III trial comparing once a day radiotherapy (QD RT) superior to cyclophosphamide, epirubicin, and vincristine
or twice a day radiotherapy (BID RT) in limited stage small regimen in small-cell lung cancer: results from a random-
cell lung cancer (LSCLC). Proc Am Soc Clin Oncol 21 ized phase III trial with 5 years’ follow-up. J Clin Oncol
(Abstract 2536) 20:4665–4672
Schiller JH, Adak S, Cella D, DeVore RF 3rd, Johnson DH Takada M, Fukuoka M, Kawahara M, Sugiiura T, Yokoyama A,
(2001) Topotecan versus observation after cisplatin plus Yokota S, Nishiwaki Y, Watanabe K, Noda K, Tamura T,
504 B. Jeremić et al.

Fukuda H, Saijo N (2002) Phase III study of concurrent (2010a) Once-daily radiotherapy to [ or = 59.4 Gy versus
versus sequential thoracic radiotherapy in combination with twice-daily radiotherapy to[or = 45.0 Gy with concurrent
cisplatin and etoposide for limited-stage small-cell lung chemotherapy for limited-stage small-cell lung cancer: a
cancer: results of the Japan clinical oncology group study comparative analysis of toxicities and outcomes. Jpn J Radiol
9104. J Clin Oncol 20:3054–3060 28:340–348
Thatcher N, Girling DJ, Howood P, Sambrook RJ, Qian W, Watkins JM, Wahlquist AE, Zauls AJ, Shirai K, Garrett-Mayer E,
Stephens RJ (2000) Improving survival without reducing Aguero EG, Silvestri GA, Sherman CA, Sharma AK (2010b)
quality of life in small-cell lung cancer patients by Involved-field radiotherapy with concurrent chemotherapy
increasing the dose-intensity of chemotherapy with granu- for limited-stage small-cell lung cancer: disease control,
locyte colony-stimulating factor support: results of a British patterns of failure and survival. J Med Imaging Radiat Oncol
medical research council multicenter randomised trial. 54:483–489
J Clin Oncol 18:395–404 White JE, Chen T, McCracken J, Kennedy P, Seydel HG,
Travis WD, Brambilla E, Muller-Hermlink HK, Harris CC Hartman G, Mira J, Khan M, Durrance FY, Skinner O
(2004) World health organziation classification of tumours. (1982) The influence of radiation therapy quality control on
Pathology and genetics of tumours of the lung, pleura, survival, response, and sites in oat cell carcinoma of the
thymus and heart. IARC Press, Lyon lung. Preliminary report of a southwest oncology group
Turrisi AT, Kim K, Blum R, Sause WT, Livingston RB, study. Cancer 50:1084–1090
Komaki R, Wagner H, Aisner S, Johnson DH (1999) Woo IS, Park YS, Kwon SH, Park YL, Lee JA, Park MJ, Hyun IG,
Twice-daily compared with once-daily thoracic radio- Jung KS, Bae HS, Oh DH, Kim WS, Park K, Park CH, Kim
therapy in limited small-cell lung cancer treated concur- HJ, Ahn YC (2000) A phase II study of VP-16-ifosfamide-
rently with cisplatin and etoposide. N Engl J Med cisplatin combination chemotherapy plus early concurrent
340:264–271 thoracic irradiation for previously untreated limited small cell
Videtic GMM, Truong PT, Dar AR, Yu EW, Stitt LW (2003) lung cancer. Jpn J Clin Oncol 30:542–546
Shifting from hypofractionated to ‘‘conventionally’’ frac- Woods RL, Levi JA (1984) Chemotherapy for small cell lung
tionated thoracic radiotherapy: a single institutions’s 10- cancer (SCLC): A randomised study of maintenance
year experience in the management of limited-stage small- therapy with cyclophosphamide, adriamycin and vincris-
cell lung cancer using concurrent chemoradiation. Int J tine (CAV) after remission induction with cis-platinum
Radiat Oncol Biol Phys 57:709–716 (CIS-DDP), VP 16–213 and radiotherapy. Proc Am Soc
Videtic GM, Belderbos JS, Spring Kong FM, Kepka L, Clin Oncol 3:214
Martel MK, Jeremic B (2008) Report from the international Work E, Nielsen O, Bentzen S, Fode K, Palshof T (1997)
atomic energy agency (IAEA) consultants’ meeting on Randomized study of initial versus late chest irradiation
elective nodal irradiation in lung cancer: small-cell lung combined with chemotherapy in limited-stage small cell
cancer (SCLC). Int J Radiat Oncol Biol Phys 72:327–334 lung cancer. J Clin Oncol 15:3030–3037
Warde P, Payne D (1992) Does thoracic radiation improve Yilmaz U, Anar C, Korkmaz E, Yapicioglu S, Karadogan I,
survival and local control in limited-stage small cell Ozkök S (2010) Carboplatin and etoposide followed
carcinoma of the lung? J Clin Oncol 10:890–895 by once-daily thoracic radiotherapy in limited disease
Watkins JM, Fortney JA, Wahlquist AE, Shirai K, Garrett- small-cell lung cancer: unsatisfactory results. Tumori 96:
Mayer E, Aguero EG, Sherman CA, Turrisi AT 3rd, Sharma AK 234–240
Radiation Therapy in Extensive Disease
Small Cell Lung Cancer
Branislav Jeremić and Luhua Wang

Contents Abstract
Approximately two-thirds of all patients with
1 Introduction.............................................................. 505 small-cell lung cancer have the disease that spread
2 Trial of Radiation Therapy in Extensive Disease beyond confines of the thorax, including as well
Small Cell Lung Cancer ......................................... 506 patients whose disease has traditionally been
3 Other Existing Studies ............................................ 509 described as ‘‘too large to be encompassed with a
tolerable radiation port’’. For these patients, che-
4 Ongoing Studies ....................................................... 510
motherapy has been considered standard treatment
References.......................................................................... 510 option for many decades. However, patterns of
failure after chemotherapy alone in this disease
reveals substantial percentage of patients failing in
the chest. It is, therefore, that curative, high-dose
thoracic radiation therapy could offer improvement
in chest disease control and, hence, improved
survival. However, in spite of these observations,
thoracic radiation therapy has mostly been used in
palliative setting. In a landmark trial, Jeremic et al
have shown that in a suitable patients, those with
best prognosis, radical chest radiation therapy can
indeed offer an improvement in local control and
overall survival over that obtained with the same
chemotherapy given alone. This was achieved at
the expense of low toxicity. recent single-institu-
tional studies from Canada and China seem to
confirm this, while two ongoing prospective stud-
ies should help further refine radiation therapy
approach in this disease.

B. Jeremić (&)
Institute of Lung Diseases, Sremska Kamenica, Serbia 1 Introduction
e-mail: nebareje@gmail.com
L. Wang For decades, clinicians and investigators considered
Department of Radiation Oncology,
chemotherapy as the standard treatment option for
Chinese Academy of Medical Sciences,
Beijing, China patients with extensive disease small cell lung cancer.

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 505
DOI: 10.1007/174_2011_208, Ó Springer-Verlag Berlin Heidelberg 2011
506 B. Jeremić and L. Wang

Given alone, it can offer the median survival time of thoracic radiation therapy (Bunn and Ihde 1981). It
9–12 months and 5-year survivals of 1–3% (Bunn should be clearly noted, however, that the majority of
et al. 1977; Beck et al. 1988; Jeremic et al. 1999). studies from that report were performed and pub-
While up to 90% of patients eventually experience lished in 1960s and 1970s. Therefore, major charac-
objective response following initial courses of che- teristics of radiation therapy course such as total
motherapy, extensive disease small cell lung cancer tumor dose, dose per fraction, and timing as well as
remains the disease with very poor prognosis. This is rather primitive treatment planning cannot be con-
so because most of patients unfortunately relapse; sidered as the optimally defining modern thoracic
leading to outcomes virtually unchanged since che- radiation therapy today. Not to be forgotten, also, is
motherapy based on platinum-etoposide combination that when one attempts to explore the effectiveness of
was introduced several decades ago. It is not hard, thoracic radiation therapy in extensive disease small
therefore, to see this disease as one of the most cell lung cancer, it must be taken into account the
frustrating challenges in thoracic oncology. To com- systemic character of extensive disease small cell
bat poor prognosis in patients with this disease when lung cancer (Ou et al. 2009). It may obscure possible
treated with chemotherapy alone, various approaches effects of thoracic radiation therapy on survival
aiming intensification of the treatment were attemp- (established on a local level), especially in adequately
ted. Unfortunately, maintenance chemotherapy after chosen subgroup of patients suitable for ‘curative’
4–6 cycles of initial chemotherapy (Splinter 1989; role of thoracic radiation therapy. Simply said,
Bunn 1992; Schiller et al. 2001) and higher doses of patients with extensive disease small cell lung cancer
chemotherapy (Ihde et al. 1994; Leyvraz et al. 2008) may have systemic progression so fast that any pos-
did not prove to be beneficial in this setting. Accu- sible effect on local control and, subsequently, sur-
mulated evidence of patterns of failure in patients vival may not be observed due to short life span of
with extensive disease small cell lung cancer treated these patients. Other issues concerning radiation
with chemotherapy alone shows that besides distant therapy, like irradiation to sites of systemic tumor
progression, local progression remains very frequent metastasis or the role of prophylactic cranial irradia-
event. Brain relapses, similarly to limited disease, are tion, were also controversial until recent EORTC
frequent event, too. It is therefore that thoracic radi- study confirmed effectiveness of prophylactic cranial
ation therapy and/or prophylactic cranial irradiation irradiation in these patients (Slotman et al. 2007).
could be of a benefit in suitable patients with exten- Trying to focus on the issues of possible
sive disease small cell lung cancer. Those would improvement in local (intrathoracic) tumor control
likely be the ones who experience some form of and its subsequent impact, if any, on overall survival
response to chemotherapy, preferably complete in favorable patient population, the role of thoracic
response as to enable meaningful prolongation of life, radiation therapy was evaluated in a prospective
which in turn, should allow enough observation time randomized trial designed in late 1987 which con-
to prove beneficial effects of either thoracic radiation tinued from 1988–1993 (Jeremic et al. 1999).
therapy and/or prophylactic cranial irradiation.
Although the place and the role of thoracic radia-
tion therapy in limited disease small cell lung cancer 2 Trial of Radiation Therapy
are well established (Murray et al. 1993; Jeremic et al. in Extensive Disease Small Cell
1997; Takada et al. 2002), the usefulness of thoracic Lung Cancer
radiation therapy in extensive disease small cell lung
cancer is still open to debate. Almost 30 years ago, in Included in this trial were treatment-na patients with
a large retrospective review of literature thoracic biopsy-proven extensive disease small cell lung can-
radiation therapy was shown to be able to reduce the cer defined as the tumor beyond the confines of the
frequency of initial chest failure. Nevertheless, com- hemithorax, mediastinum, and ipsilateral or contra-
plete response rates, overall response rates, the med- lateral supraclavicular nodes. Patients with tumors
ian survival time, and 2-year disease-free survival that could not be encompassed within a tolerable
were identical for patients treated with chemotherapy thoracic radiation therapy field as well as those having
alone and those treated with chemotherapy and an ‘isolated’ pleural effusion with positive cytology
Radiation Therapy in Extensive Disease Small Cell Lung Cancer 507

were also considered as having extensive disease until death (treated with supportive care only) or
small cell lung cancer. Patients with negative cytol- treated with oral etoposide, 50 mg/m2, days 1–21,
ogy in an ‘isolated’ pleural effusion were considered every 28 days to a total of six cycles or further until
ineligible for this study. Other eligibility criteria further progression.
included a Karnofsky performance status score of Thoracic radiation therapy was performed using
C70, age 18–70 years, and adequate haematological, 6–10 MV photons from linear accelerators. Target
renal, and hepatic function (unless due to liver volume included all gross disease and ipsilateral
metastases). Excluded were patients with recent or hilum with a 2 cm margin, and the entire mediasti-
concurrent severe, uncontrolled cardiovascular or num with a 1 cm margin and both supraclavicular
pulmonary disease as well as those with central ner- fosse were routinely irradiated. Anteroposterior-pos-
vous system metastases or other abnormality when teroanterior treatment fields were used to deliver
substantially impairing mental status. For staging 36 Gy in 24 fractions in 12 treatment days over
purposes chest X-rays and tomography, bronchos- 2.5 weeks, after which various combinations of
copy, bone marrow biopsy, brain, bone and liver treatment fields were used to give additional 18 Gy in
radionuclide scans, and abdominal ultrasonography 12 fractions in six treatment days. Therefore, the total
were done. Computerized tomography scans of the tumor dose was 54 Gy in 36 fractions in 18 treatment
thorax, brain, and abdomen were highly recom- days in 3.5 weeks. Normal tissue dose limits included
mended as well as pulmonary function tests and the maximum dose of 36 Gy to the spinal cord and the
actually became mandatory in all patients treated entire heart, 54 Gy for the oesophagus, and 18 Gy for
from 1989. Treatment started with three cycles of the contralateral lung. Two daily fractions of 1.5 Gy
standard-dose cisplatin-etoposide regimen given at were used. During accelerated hyperfractionated
3-week intervals: cisplatin, 80 mg/m2, day 1, and radiation therapy, on each thoracic radiation therapy
etoposide, 80 mg/m2, days 1–3. After this portion of treatment day 50 mg of carboplatin and 50 mg of
the treatment schedule, complete patient reevaluation etoposide were both given between the two daily
and restaging were performed, using the procedures fractions (3–4 h after the first one, i.e. 1–2 h before
outlined above. Patients achieving complete response the second one). Prophylactic cranial irradiation was
at local and distant levels and those achieving partial administered with tumor dose 25 Gy in ten daily
response within the thorax accompanied with the fractions in 2 weeks to the whole brain via two par-
complete response elsewhere were then randomized allel—opposed lateral fields in groups I and II.
to receive either (a) accelerated hyperfractionated Patients in groups III and IV also received prophy-
radiation therapy and concurrent low-dose daily lactic cranial irradiation, but only in cases achieving
chemotherapy consisting of carboplatin and etoposide complete response at distant level. When appropriate,
50 mg each, given on each radiation therapy treat- palliative radiation therapy with 30 Gy in ten daily
ment day, followed by prophylactic cranial irradiation fractions in 2 weeks was offered to patients with
and then by additional two cycles of cisplatin-etopo- metastatic lesions. Follow-up consisted of the fol-
side (group I) or (b) four additional cycles of cis- lowing: patients were fully examined at the end of
platin-etoposide and prophylactic cranial irradiation their treatment (groups I–IV), every month for
(group II). Remaining patients (achieving worse 6 months after the end of the treatment,
response) were not a part of randomization procedure. every 2 months for 2 years thereafter, and every
Patients achieving worse response, i.e. those achiev- 4–6 months thereafter. By using the diagnostic tools
ing complete response or partial response within outlined above restaging was made at time of pro-
thorax, but only a partial response elsewhere (group gression. Patients were evaluated for response at the
III and group IV), were offered two additional cis- prespecified time points: (1) after three cycles of
platin-etoposide cycles followed by the same accel- cisplatin-etoposide (week 9), then (2) after either
erated hyperfractionated radiation therapy/ accelerated hyperfractionated radiation therapy or two
carboplatin-etoposide and in case of complete additional cisplatin-etoposide cycles (week 15), and
response at distant level, also prophylactic cranial (3) at the end of treatment (week 21). The Eastern
irradiation. Patients achieving either stable disease or Cooperative Oncology Group and the Radiation
progressive disease (group V) were either observed Therapy Oncology Group/European Organization for
508 B. Jeremić and L. Wang

the Research and Treatment of Cancer toxicity criteria radiation therapy/carboplatin-etoposide (group I) or
were used in combination to address the issue of two additional cycles of PE (group II) were admin-
chemotherapy- and radiation therapy-induced istered), the complete response rate was significantly
toxicity. higher in group I than in group II (96 vs. 61%,
A total of 210 patients entered this study. Four p = 0.000007), and it persisted until week 21 when
patients were excluded from analysis due to various actual response rates for the groups I and II were 96
reasons, making, therefore a total of 206 patients fully and 66%, respectively (p = 0.00005). Looking at
evaluable for toxicity and survival. There was no both absolute increase in percent responders and the
difference in the distribution of various variables tempo of its achievement, the 4th and the 5th cycles
between the five treatment groups. For all 206 of chemotherapy add nothing to the response
patients, the median survival time was 9 months, and achieved in group I patients after accelerated hyper-
survival rates at 1, 2, 3, 4, and 5 years were 38, 19, fractionated radiation therapy had been added to three
9.7, 4.9 and 3.4%, respectively. Further data and the cycles of cisplatin-etoposide. Furthermore, the 6th
discussion are limited to patients in the randomized and 7th cycles of cisplatin-etoposide in the chemo-
part of the study (groups I and II). therapy alone group brought only a few percent
Patients in group I (radiochemotherapy) achieved increase in response rates. Therefore, it seems that
the results that were significantly better than those in after three cycles of induction chemotherapy followed
group II (chemotherapy alone): the median survival by thoracic radiation therapy, no additional gain was
time was 17 vs. 11 months (p = 0.041), and 5-year observed with additional chemotherapy in the group
survival rates were 9.1 and 3.7% for groups I and II, (I). Similarly, addition of 6th and 7th cycle of che-
respectively. Local recurrence-free survival was also motherapy in chemotherapy alone group (II) added no
better in group I than in group II, with median time to visible gain in response rates. These data altogether
local recurrence of 30 and 22 months, respectively, question the duration (number of cycles) of chemo-
and 5-year local recurrence-free survival of 20 and therapy as it is practiced nowadays. They also set up
8.1%, respectively (p = 0.062). Distant metastasis- the stage and scene for possible clinical trial in the
free survival was similar between the two groups. future using standard-dose chemotherapy (e.g., cis-
Although group II patients treated with chemotherapy platin-etoposide) with or without thoracic radiation
only achieved longer median time to distant metas- therapy.
tasis than group I patients treated with combined Haematological high-grade ([3) toxicity (leuco-
chemotherapy/thoracic radiation therapy (16 vs. poenia, thrombocytopenia, and anaemia) was more
14 months, respectively), they had 5-year distant frequent in chemotherapy-only (group II) than in
metastasis-free survival half of that observed in the radiochemotherapy (group I), though not significant.
combined group (14 vs. 27%), the difference being There was no difference between groups I and II
insignificant (p = 0.35). Because local recurrence- regarding incidence of high-grade infection
free survival rate was only marginally insignificant (p = 0.64). Nausea and vomiting were significantly
and distant metastasis-free survival rate was not sig- more frequent in group II than in group I
nificantly different between groups I and II, first (p = 0.0038), due to more cycles of chemotherapy
relapse-free survival analysis was done next and administered to patients in group II. Similar was
showed significant superiority of patients in group I observed in case of alopecia (p = 0.000003). High-
(median time to first relapse, 13 vs. 9 months, grade kidney toxicity was observed only in group II.
respectively; 1–5 year first relapse-free survival rate; Acute high-grade (C3) radiation therapy-induced
(p = 0.045)). oesophagitis was observed only in patients who
Analysis of response rates provided the local received thoracic radiation therapy. Due to a few
complete response rates in groups I and II at weeks 9, cases of radiation therapy-induced high-grade bron-
15, and 21. At week 9 (i.e., after three cycles of chopulmonary toxicity, the difference between the
induction cisplatin-etoposide, before randomization), two groups was not significant (p = 0.082).
there was no difference between the two groups in the Study of Jeremic et al. (1999) was the very first
local response rate (47 vs. 44%, p = 0.77). At prospective randomized study that evaluated curative
week 15 (when either accelerated hyperfractionated thoracic radiation therapy in extensive disease small
Radiation Therapy in Extensive Disease Small Cell Lung Cancer 509

cell lung cancer. It showed that thoracic radiation therapy with no delays. One patient received one
therapy may have an important place and may have a cycle of chemotherapy, three received three cycles
substantial role in overall treatment of patients with and 28 received four cycles. There were four com-
extensive disease small cell lung cancer. In an effort plete and 28 partial responses to chemotherapy. All
to gain more insight in the study results and perhaps study patients received prophylactic cranial irradia-
additional information for future studies, a multivar- tion. The median time to disease progression was
iate analysis of the most common pretreatment 8.4 months and the median overall survival time was
prognostic factors in these patients was performed. 13.7 months. There have been 13 distant-only recur-
Karnofsky performance status score and weight loss rences and six combined distant-local recurrences.
were strong prognosticators of improved treatment There were no treatment-related deaths. Maximal
outcome. Of particular importance, the number of radiation therapy-induced toxicity was grade 2
metastases significantly and independently predicted oesophagitis in 18 patients which resolved in all
improved overall survival. Patients with only one patients.
metastasis had better outcome than those with C2 In another study, also only preliminarily reported
metastases, showing that metastatic tumor burden (Zhu et al. 2010), of a total of 154 patients 89 patients
should be taken into account in future studies. Finally, received combined radiochemotherapy and 65
overall good results should be attributed, at least in a patients received chemotherapy alone. Chemotherapy
part, to the fact that approximately 90% of all patients regimens were either cisplatin-etoposide or carbo-
in that study had 1–2 metastases. It is therefore that platin-etoposide. Thoracic radiation therapy dose
subsequent discussion of the study results including ranged 40–60 Gy given in 1.8–2.0 Gy per fraction.
prognostic factors analysis was frequently done using No prophylactic cranial irradiation was planned for
the term limited extensive disease trying to emphasize this patient population as it was not standard treat-
low tumor burden in these patients. ment option in the study period (January 2003–
December 2006). For the whole group, the medians
survival time was 13.7 months and the 2- and 5-year
3 Other Existing Studies survival rates were 27.9 and 8.1%, respectively.
Corresponding figures for chemotherapy-alone group
Recently, preliminary data from two studies became were 9.3 months, 16.9 and 4.6%, respectively, while
available. In a Canadian trial, Yee et al. (2010) found for radiochemotherapy group were 17.2 months, 36
eligible patients had biopsy-proven extensive disease and 10.1%, respectively (p = 0.0001). The incidence
small cell lung cancer and attained an objective of thoracic failures in two treatment groups was 70 vs.
radiologic disease response after receiving at least one 29.6% (p \ 0.001). What these two studies have
chemotherapy cycle. Study patients were also offered shown is, albeit of some shortcomings, that thoracic
prophylactic cranial irradiation (25 Gy in ten daily radiation therapy indeed can decrease the incidence of
fractions in 2 weeks) which was given simultaneously chest failures and leads to promising survival data. In
with thoracic radiation therapy (40 Gy in 15 daily addition, as an important factor in understanding
fractions in 3 weeks) 4–6 weeks after completing therapeutic benefit, toxicity of combined radioche-
chemotherapy. Target volume in thoracic radiation motherapy in patients with extensive disease small
therapy was post-chemotherapy residual disease and a cell lung cancer was low.
margin. Endpoints in this study included overall sur- Studies of Zhu et al. (2010) and Yee et al. (2010)
vival, disease-free survival, local control, and toxic- should also not only be seen as a confirmatory data of
ity. Thirty-two of 33 accrued patients were evaluable. the study of Jeremic et al. (1999) but also as confir-
Seventeen patients had \3 metastases, while 2 matory of existing institutional practices among
patients had at least three metastases. Bulky intra- radiation oncologists and medical oncologists
thoracic disease was present alone in 3 patients, and it involved in the treatment of extensive disease small
was accompanied by \3 metastases in 1 patient. cell lung cancer since the time study of Jeremic et al.
Pleural/pericardial disease was alone present in 3 (1999). This was recently brought to the evidence by
patients and was accompanied with \3 metastases in the study of Ou et al. (2009) who retrospectively
6 patients. All but three patients completed radiation analyzed the data from the Cancer Surveillance
510 B. Jeremić and L. Wang

programs of Orange, San Diego, and Imperial coun- Table 1 Basic comparison RTOG 0835 vs. CREST
tries in southern California covering an area with Trial Nature Diagnosis Patients Sequencing
estimated population of 6.2 million. Small cell lung (classic/ (n mets) Rt-chemo
cancer patients diagnosed between 1991 and 2005 pet)
who had complete follow-up data were included in RTOG Curative More More Sequential
the study. Extensive disease was defined as distant precise favorable
disease stage according to Surveillance, Epidemiol- CREST Palliative Less Less Sequential
ogy, and end results summary staging. There were precise favorable
3,428 such patients. Of them, radiation therapy was
given in 1,204 (35.1%) patients. A number of
with brain restaging done and with 0–1 residual sites
clinicopathologic characteristics were analyzed upon
of extrathoracic disease present at the time of re-
their influence on the outcome. The 1-year, 2-year,
staging. Radiotherapy part of the study includes tho-
and median overall survival of extensive disease
racic radiation therapy dose of 45 Gy in 15 fractions,
small cell lung cancer patients who received radiation
prophylactic cranial irradiation of 25 Gy in 10 frac-
therapy were 27.8, 9.3%, and 8 months and were
tions, while 45 Gy in 15 fractions will be given to
significantly better than extensive disease small
metastatic lesions. Major objectives of the trial
cell lung cancer patients who did not receive
included (1) overall median and 1-year survival (2)
radiation therapy (16.2, 3.8%, and 4 months, respec-
recurrence patterns and time to failure, as well as (3)
tively; p \ 0.0001). Cox multivariate analysis of
acute and late toxicity. Similarly, the Dutch Lung
potential prognostic factors confirmed independent
Cancer Study Group is executing a chest radiotherapy
positive influence of delivered radiation therapy on
in extensive disease small cell lung cancer trial
treatment outcome (HR, 0721; 95% CI, 0-670-0.776;
(CREST) with the primary endpoint being overall
p \ 0.001). Unfortunately, other important charac-
survival. Secondary endpoints include pattern of
teristics of radiation therapy (e.g., curative vs. palli-
relapse and toxicity. In CREST trial, patients with
ative, time-dose-fractionation patterns, sequencing of
extensive disease small cell lung cancer without brain
chemotherapy and radiation therapy) were not pro-
metastasis or pleural metastasis undergo chemother-
vided as to enable better insight into patterns of
apy. Those achieving any response to 4–6 cycles of
practice of radiation therapy in this disease. Never-
chemotherapy are randomized to prophylactic cranial
theless, these data suggest that radiation therapy is
irradiation and no thoracic radiation therapy versus
practiced in about 1/3 of these patients improving
those treated with prophylactic cranial irradiation and
survival. These may be a good starting point for more
thoracic radiation therapy (30 Gy in 10 fractions)
practicing in the near future, especially once we have
given only if the toxicity of the required fields will not
more solid data from ongoing trials.
be prohibitive. These two trials are different in many
characteristics, and some of major strategic charac-
teristics are briefly outlined in a Table 1. It is
4 Ongoing Studies expected that these two studies will bring additional
substance to the data of Jeremic et al. (1999) and help
After a gap of almost 10 years following the publi-
optimize both treatment approach with thoracic radi-
cation of this landmark study (i.e., 20 years since its
ation therapy and identification of suitable patients for
start!), investigators over the world finally started
thoracic radiation therapy.
with preparations for additional prospective random-
ized trials of thoracic radiation therapy in extensive
disease small cell lung cancer. The Radiation Therapy
References
Oncology Group in the US planned and opened up a
study (RTOG 0835) in which patients with extensive
Beck LK, Kane MA, Bunn PA Jr (1988) Innovative and future
disease small cell lung cancer and no brain metastasis, approaches to small cell lung cancer treatment. Semin
having Eastern Cooperative Oncology Group perfor- Oncol 15:300–314
mance status 0–2 are enrolled. Patients would have to Bunn PA Jr (1992) Clinical experience with carbolatin
achieve either complete response or partial response, (paraplatin) in lung cancer. Semin Oncol 19(suppl 2):1–11
Radiation Therapy in Extensive Disease Small Cell Lung Cancer 511

Bunn PA, Ihde DC (1981) Small cell bronchogenic carcinoma: treatment of limited stage small cell lung cancer. J Clin
a review of therapeutic results. In: Liv-ingston RB (ed) Oncol 11:336–344
Lung Cancer. Boston, Martin Nijhoff, pp 169–208 Ou S-H, Ziogas A, Zell JA (2009) Prognostic factors for
Bunn PA Jr, Cohen MH, Ihde DC, Fossieck BE Jr, Matthews survival in extensive stage small-cell lung cancer (ED-
MJ, Minna JD (1977) Advances in small cell bronchogenic SCLC). The importance of smoking history, socioeconomic
carcinoma: a commentary. Cancer Treat Rep 61: and marital statuses, and ethnicity. J Thorac Oncol 4:37–43
333–342 Schiller JH, Adak S, Cella D, DeVore RF 3rd, Johnson DH
Ihde DC, Mulshine JL, Kramer BS, Steinberg SM, Linnoila RI, (2001) Topotecan versus observation after cisplatin plus
Gazdar AF, Edison M, Phelps RM, Lesar M, Phares JC etoposide in extensive-stage small-cell lung cancer: E7593–
(1994) Prospective randomized comparison of high-dose a phase III trial of the Eastern cooperative oncology group.
and standard-dose etoposide and cisplatin chemotherapy in J Clin Oncol 19:2114–2122
patients with extensive-stage small cell lung cancer. J Clin Slotman B, Faivre-Finn C, Kramer G, Rankin E, Snee M,
Oncol 12:2022–2034 Hatton M, Postmus P, Collette L, Musat E, Senan S (2007)
Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1997) EORTC Radiation oncology group lung cancer group
Initial versus delayed accelerated hyperfractionated radia- prophylactic cranial irradiation in extensive small-cell lung
tion therapy and concurrent chemotherapy in limited small cancer. N Engl J Med 357:664–672
cell lung cancer. J Clin Oncol 15:893–900 Splinter TAW (1989) Chemotherapy of small cell lung cancer
Jeremic B, Shibamoto Y, Nikolic N, Milicic B, Milisavljevic S, (SCLC): duration of treatment. Lung Cancer 5:186–196
Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999) Takada M, Fukuoka M, Kawahara M, Sugiura T, Yokoyama A,
The role of radiation therapy in the combined modality Yokota S, Nishiwaki Y, Watanabe K, Noda K, Tamura T,
treatment of patients with extensive disease small-cell lung Fukuda H, Saijo N (2002) Phase III study of concurrent
cancer (ED SCLC): a randomized study. J Clin Oncol versus sequential thoracic radiotherapy in combination with
17:2092–2099 cisplatin and etoposide for limited-stage small-cell lung
Leyvraz S, Pampallona S, Martinelli G, Ploner F, Perey L, cancer: results of the Japan clinical oncology group study
Aversa S, Peters S, Brunsvig P, Montes A, Lange A, Yilmaz 9104. J Clin Oncol 20:3054–3060
U, Rosti G (2008) Solid tumors working party of the Yee D, Butts C, Chu Q (2010) Phase II trial of consolidation
European group for blood and marrow transplantation. A chest radiotherapy for extensive stage small cell lung cancer
threefold dose intensity treatment with ifosfamide, carbo- [Abstract]. Radiother Oncol 96(1):102
platin, and etoposide for patients with small cell lung Zhu H, Zhou Z, Wang Y, Bi N, Feng Q, Li J, Jima LV, Chen D,
cancer: a randomized trial. J Natl Cancer Inst 100:533–541 Shi Y, Wang L (2010) Thoracic radiation therapy improves
Murray N, Coy Pater J, Hodson I, Arnold A, Zee BC, Payne D, the overall survival of patients with extensive disease small
Kostashuk EC, Evans WK, Dixon P (1993) Importance of cell lung cancer with distant metastasis. proc. Am Soc Ther
timing for thoracic irradiation in the combined modality Oncol Radiol 72, Abstract 2211
Prophylactic Cranial Irradiation in Small-Cell
Lung Cancer
Michael C. Stauder and Yolanda I. Garces

Contents Abstract
Prophylactic cranial radiation (PCI) has been used in
1 Introduction.............................................................. 514 the management of small-cell lung cancer given the
2 Studies Evaluating Prophylactic Cranial propensity of disease relapse in the brain. Several
Irradiation ................................................................ 514 studies have shown that PCI reduces the rate of brain
2.1 Retrospective Studies ................................................ 514 metastases and improves survival. However, the use
2.2 Randomized Trials..................................................... 514
of PCI in the management of small-cell lung cancer
2.3 Meta-Analyses ........................................................... 515
2.4 Extensive Stage Small-Cell Lung Cancer ................ 516 has been controversial due to conflicting evidence of
efficacy and toxicities. For example, the most
3 Treatment Schedule................................................. 516
3.1 Dose and Fractionation ............................................. 516 effective radiation dose and fractionation regimen
3.2 Timing of PCI............................................................ 516 is not known. Also, conflicting reports on the
neurotoxicity of PCI make it difficult to make
4 Neurotoxicity and Quality of Life ......................... 517
4.1 Retrospective Studies ................................................ 517 definitive conclusions regarding the side-effects of
4.2 Prospective Trials ...................................................... 517 treatment. As the survival of patients with SCLC
4.3 Extensive Stage Small-Cell Lung Cancer ................ 518 improves, the controversies regarding the neuro-
5 Patterns of Care....................................................... 518 toxic effect of PCI may become more defined.
6 Future Directions..................................................... 519
Certainly, well-designed prospective clinical
trials addressing these issues are needed and are
7 Conclusions ............................................................... 519
ongoing.
References.......................................................................... 519

Abbreviations

Gy Gray
Fx Fractions
BID Twice-daily
LS Limited stage
ES Extensive stage
SCLC Small-cell lung cancer
NSCLC Non-small-cell lung cancer
M. C. Stauder  Y. I. Garces (&)
Radiation Oncology, Mayo Clinic College of Medicine,
Charlton Building, 200 First Street SW, Rochester,
MN 55905, USA
e-mail: garces.yolanda@mayo.edu

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 513
DOI: 10.1007/174_2011_270, Ó Springer-Verlag Berlin Heidelberg 2011
514 M. C. Stauder and Y. I. Garces

Similarly, other trials from the 1970s also con-


1 Introduction cluded that PCI decreased the incidence of brain
metastases but did not improve overall survival
Small-cell lung cancer (SCLC) comprises 15–25% of (Jackson et al. 1977; Cox et al. 1978). However, when
all lung cancer cases. A complete response to treat- restricted to patients experiencing a complete
ment can be seen in 50–80% of patients undergoing response to chemoradiotherapy, other more modern
chemoradiotherapy for limited stage disease (Turrisi trials suggest that there may indeed be a survival
et al. 1999; Levitan et al. 2000; Bremnes et al. 2001), benefit with the use of PCI (Ohonoshi et al. 1993;
and in 40–50% of patients with extensive disease Shaw et al. 1994; Arriagada et al. 1995; Gregor et al.
(Jeremic et al. 1999). Unfortunately, disease relapse 1997; Laplanche et al. 1998). Several randomized
within the central nervous system (CNS) is common. trials have been performed in an attempt to better
Radiotherapy directed at the brain for prevention of define the role of PCI in the management of SCLC.
CNS relapse, or prophylactic cranial irradiation (PCI) Several of these trials are listed in Table 1.
has been used in an attempt to decrease the incidence
of brain metastasis. However, the use of PCI in the
management of small-cell lung cancer has been con- 2.2 Randomized Trials
troversial due to conflicting evidence of efficacy and
toxicities. SCLC has a propensity to metastasize to A French prospective randomized trial (PCI-85) was
the brain and incidence of brain metastasis can be up performed to evaluate the effect of PCI on the inci-
to 15% at diagnosis. Also, approximately 50–70% of dence of brain metastases, overall survival, and late
small-cell lung cancer patients who do not receive toxic effects of treatment. A total of 300 patients with
PCI will ultimately develop clinical evidence of brain SCLC in complete remission were enrolled at 21
metastases. Autopsy series report an even higher centers from March 1985 to March 1993. Patients
incidence of 80% at 2 years (Nugent et al. 1979). The with extensive stage SCLC comprised 20% of the
introduction of PCI in SCLC was first proposed in sample population. Randomization was done to PCI at
1973 by (Hansen et al. 1973) due to the dissatisfaction a dose of 24 Gy/8 Fx (n = 149) or no PCI (n = 151).
with the high rate of brain metastasis following pro- PCI was given at the time of determination of com-
longed survival after the use of novel chemothera- plete remission and no chemotherapy was allowed
peutic agents in patients with limited and/or extensive during PCI or one week before and after (Arriagada
stage disease. et al. 1995). The incidence of brain metastasis at
2-years was decreased in the arm receiving PCI
(40 vs. 67%, p \ 10-13) which translated into a non-
2 Studies Evaluating Prophylactic statistically significant improvement in 2-year overall
Cranial Irradiation survival (29 vs. 21.5%, p = 0.14). A follow-up study
(PCI-88) was closed early based on the interim
2.1 Retrospective Studies analysis showing such a significant decrease in the
incidence of brain metastasis that most investigators
One of the early retrospective studies evaluating the felt that PCI should be administered to all patients
role of PCI in the management of SCLC was per- (Laplanche et al. 1998).
formed in the mid 1970s by Komaki and colleagues. A Japanese trial evaluating 46 patients with SCLC
A total of 131 patients were analyzed for the inci- in complete remission showed a significant decrease
dence of brain metastasis of patients when PCI was in the rate of brain metastasis with the use of PCI
included as part of their small-cell lung cancer ther- (Ohonoshi et al. 1993). The incidence of CNS relapse
apy. Of these patients, 57 received PCI and 74 in the group receiving PCI was 22 versus 52% in
patients did not. The incidence of CNS failure at 12 those where it was omitted. A trend for an improved
and 24 months was significantly decreased in the median overall survival (21 vs. 15 months) was also
patients receiving PCI versus those not receiving PCI seen in the group receiving PCI. The 5-year overall
(11 vs. 28%, and 11 vs. 58%, respectively; p \ 0.01) survival rate was 22 and 13% in the patients where
(Komaki et al. 1981). PCI was delivered and not delivered, respectively.
Prophylactic Cranial Irradiation in Small-Cell Lung Cancer 515

Table 1 Characteristics of trials examining the role of PCI in SCLC


Brain metastasis Overall survival
Study N PCI dose* PCI No-PCI PCI No-PCI
Arriagada et al. (1995) 300 24 Gy/8 Fx 40% 67% 29% 21.5%
2 years 2 years 2 years 2 years
Laplanche et al. (1998) 211 24 Gy/8 Fx 44% 51% 22% 16%
4 years 4 years 4 years 4 years
Ohonoshi et al. (1993) 46 40 Gy/20 Fx 22% 52% 22% 13%
5 years 5 years
Gregor et al. (1997) 314 30 Gy/10 Fx 30% 38% 25% 19%
2 years 2 years 2 years 2 years
Aupérin et al. (1999) 987 24–25 Gy/8–12 Fx 33.3% 58.6% 20.7% 15.3%
3 years 3 years 3 years 3 years
Slotman et al. (2007) 286 20 Gy/5 Fx 14.6% 40.4% 27.1% 13.3%
1 year 1 year 1 year 1 year
*
Most common dose
PCI prophylactic cranial radiation, Gy Gray, Fx fractions

A three-arm trial performed by the UKCCCR and 2.3 Meta-Analyses


EORTC was designed to randomize 314 patients to
either PCI at a dose of 36 Gy/18 Fx or 24 Gy/12 Fx Given the suggestion of improved survival with PCI,
versus no PCI (Gregor et al. 1997). The trial was a meta-analysis was performed, which included 7
remarkable for a very poor accrual rate so it was trials consisting of 987 patients with limited stage
redesigned to allow the physician to choose a PCI SCLC in complete remission after primary chemora-
dose regimen. Most physicians chose 30 Gy/10 Fx diotherapy (Aupérin et al. 1999). The use of PCI
(n = 61) and 8 Gy/1 Fx (n = 25). Results from this improved the incidence of brain metastasis at 3-years
study revealed that with PCI, a significant reduction (33.3 vs. 58.6% at 3 years, p \ 0.001). An absolute
in brain metastases was seen (38 vs. 54%, HR = 0.44, 3-year overall survival benefit of 5.4% was also seen
95% CI 0.29–0.67), as well as an improvement (15.3 vs. 20.7%, p = 0.01) favoring the use of PCI.
in metastasis-free survival (HR = 0.75, 95% CI However, there have been some criticisms of the
0.58–0.96). No difference in overall survival was methodology employed in this analysis. Over half of
observed between the group receiving PCI and those the trials included (4 of 7) enrolled less than 100
not receiving PCI with a median survival of 305 days patients and approximately 15% of patients had
and 300 days, respectively. extensive stage disease. Also, the dose-fractionation
As in SCLC, the use of PCI after complete remission regimens were not uniform among the trials but as
in patients with non small-cell lung cancer (NSCLC) in the UKCCCR/EORTC trial, this analysis also
has shown to be effective in preventing brain metasta- suggests that higher doses of RT led to a greater
ses. Recently, a randomized trial by RTOG has been reduction in the incidence of brain metastasis.
reported which compared PCI versus observation in Another meta-analysis which included 12 ran-
patients with stage III NSCLC without progression domized trials and 1547 patients had a similar con-
after definitive therapy (Gore et al. 2011). The use of clusion (Meert et al. 2001). Only 5 of the 12 trials
PCI decreased the 1-year rate of brain metastasis consisted exclusively of patients known to be in
compared to the group that did not receive PCI (7.7 vs. complete remission after primary therapy. In the other
18%, respectively (p = .004)). No difference was trials, 5 included patients given PCI at the time of
observed in overall survival between groups. induction chemotherapy, and 2 included patients
516 M. C. Stauder and Y. I. Garces

given PCI at end of induction chemotheraphy without was seen among the patients in this study. Addition-
any restaging. A decrease in the incidence of meta- ally, a higher biologically equivalent dose (BED) did
static brain lesions was seen for all patients given PCI not significantly decrease the incidence of brain
(HR 0.48 (95% CI: 0.39–0.60)) and an improvement metastases. On further analysis, the 5-year overall
in overall survival was seen for patients in complete survival rate was improved in patients who received a
remission given PCI (HR 0.82; 95% CI: 0.71–0.96). BED of \39 Gy10 compared to those receiving a
BED of [39 Gy10 (22.3 vs. 13.3%, p = 0.03). It is
worthwhile to note, however, that only a small num-
2.4 Extensive Stage Small-Cell Lung ber of patients (n = 6) in this review actually
Cancer received a dose [39 Gy.
A large Intergroup trial recently attempted to address
While several randomized studies have been per- this question by enrolling 720 patients with limited
formed in limited stage SCLC to evaluate the effect of stage SCLC in a randomized trial looking at a standard
PCI on brain metastasis and survival, data is much PCI dose (25 Gy/10 Fx) versus a higher dose of PCI
more limited in extensive stage SCLC. One large (36 Gy/18 Fx or 36/24 BID Fx). Patients in this trial
randomized trial included patients with extensive needed to have a documented complete remission prior
stage SCLC and any response to 4–6 cycles of che- to inclusion. Patients from 157 different treatment
motherapy (Slotman et al. 2007). The patients in this centers in 22 different countries were represented
trial received either PCI (20/5 Fx or 30/12 Fx) or no (Le Pechoux et al. 2009). No significant difference was
radiotherapy. The results show that the cumulative seen in the 2-year incidence of brain metastasis between
risk of brain metastasis at one year was decreased the high and low-dose groups (23 and 29%, respectively
in the group that received PCI (14.8 vs. 40.4%, (p = 0.18)). Paradoxically, a decreased 5-year overall
p \ 0.001) as was the progression-free survival (14.7 survival rate was seen in the high-dose arm compared to
versus 12 weeks, respectively, p = 0.02). Although, the low-dose arm (42 vs. 37%, respectively (p = 0.05)),
not a primary end point of the trial, overall survival at but of note, survival was not the primary endpoint of the
1 year was also improved in the patients receiving trial. The authors concluded that 25 Gy in 10 fractions
PCI compared to the no radiotherapy group (27 vs. should remain the standard of care.
13%, respectively, p = 0.003). Altered fractionation has also been evaluated as a
potential method to improve control of brain metas-
tasis and promote improved survival. A single insti-
3 Treatment Schedule tution Phase II trial comparing twice-daily PCI to
observation reported a 2-year disease-free and overall
3.1 Dose and Fractionation survival of 54 and 62%, respectively in the 15 patients
who received PCI (Wolfson et al. 2001).
One of the important findings of the UKCCCR/ Based on the lack of evidence for increased effec-
EORTC study is that a suggestion of a dose response tiveness of a higher PCI dose or altered fractionation, a
was seen due to the fact that those receiving 24 Gy/12 25 Gy/10 fraction regimen, corresponding to the stan-
Fx PCI had no difference in the incidence of brain dard-dose arm of the intergroup trial, has become a
metastasis compared to those patients not receiving commonly adopted PCI regimen. The currently open
any PCI and the patients who appeared to benefit the RTOG 0937 trial examining PCI alone versus PCI and
most from PCI were those who received 36 Gy in 18 consolidative extracranial RT for extensive stage
Fx (Gregor et al. 1997). This relationship was also SCLC uses this regimen. (NCT01055197).
seen in the meta-analysis performed by the Prophy-
lactic Cranial Irradiation Overview Collaborative
Group (Aupérin et al. 1999). 3.2 Timing of PCI
Conflicting data comes from Canadian investiga-
tors who reported the results of a retrospective review The large meta-analysis conducted by Aupérin et al.
of 163 patients with limited stage SCLC given PCI (1999) suggests that control of brain metastases
(Tai et al. 2003). No difference in dosing schedule improves as the delay to receiving PCI is decreased.
Prophylactic Cranial Irradiation in Small-Cell Lung Cancer 517

On sub-group analysis, a significant reduction in the (Komaki et al. 1995). The patients analyzed had a
risk of brain metastasis was seen as the time between complete response (1 PR) to primary therapy and were
the start of induction therapy decreased. The relative administered a panel of widely used standardized
risk of developing brain metastases compared to a neuropsychological tests. On initial evaluation, 97% of
control group was 0.27 in the patients receiving patients had evidence of cognitive dysfunction prior to
PCI \ 4 months from the start of induction therapy, receiving PCI. The most common deficiencies were in
0.50 in the 4–6 month group, and 0.69 in the verbal memory, frontal lobe dysfunction, and fine
[6 month group (p = 0.01) (Aupérin et al. 1999). motor coordination. Excluding the patients with
In another study, the rate of brain metastases was underlying medical conditions such as stroke, mild
decreased in patients receiving PCI after 2–3 cycles mental retardation, learning disability , or alcohol
of chemotherapy compared to after 5–6 cycles. The abuse, 20 of 21 patients still displayed abnormal test-
group of patients having PCI delayed an average of ing. Additional testing of 11 patients at 6–20 months
170 days had a 15% rate of developing overt metas- after PCI revealed no significant difference in any of the
tases before PCI started (Lee et al. 1987). tests compared to baseline. A dose of 25 Gy delivered
A radiobiologic analysis was performed to evalu- in 10 fractions was used in all patients analyzed.
ate the effect of increased dose on brain relapse rate in A corollary study published in 2008 confirms these
patients with SCLC (Suwinski et al. 1998). Using data results (Grosshans et al. 2008). In the 17 patients with
from 42 trials which report the incidence of brain extended follow-up (mean 1.5 years), early declines in
metastases both with and without the use of PCI, these executive function and expressive language tests were
investigators calculated that in patients not receiving observed. When controlling for disease progression, no
PCI, the cumulative incidence of brain metastases was differences were seen from pre-PCI testing. Testing at
32%. In the group of patients receiving PCI within later time points ([450 days) revealed significant
60 days of starting primary therapy, a linear dose– improvements in expressive speech and motor coordi-
response was established up to a dose of 35 Gy when nation. In another study which showed improved brain
delivered daily as 2 Gy fractions. The same rela- control and survival in the group of patients treated with
tionship was not seen, however, if PCI was delayed PCI, late neurotoxicity was observed infrequently with
and delivered after 60 days. Also, in patients receiv- only 1 patient experiencing mild neurological deterio-
ing early PCI (\60 days), the dose of radiation nee- ration (Ohonoshi et al. 1993).
ded to produce a 50% reduction in the rate of brain
metastases that was less than in those receiving late
PCI (20 vs. 27 Gy, respectively). 4.2 Prospective Trials

The results regarding neuropsychological testing,


4 Neurotoxicity and Quality of Life global health status, and quality of life outcomes in
patients enrolled in randomized trials are also mixed
4.1 Retrospective Studies (Arriagada et al. 1995; Gregor et al. 1997; Slotman
et al. 2007).
In the large meta-analyses showing improved brain In the PCI-85 study, patients underwent neuro-
control and survival in the patients who received PCI, psychological testing which included temporo-spatial
the neurotoxicity experienced is not well described and orientation, memory, judgment, language, praxis, and
data regarding the extent of neurotoxicity induced by mood status (Arriagada et al. 1995). The tests were
PCI is inconclusive (Aupérin et al. 1999; Meert et al. performed at randomization and at 6, 18, 30, and
2001). Data from the Mayo Clinic and North Central 48 months later if neurological symptoms appeared.
Cancer Treatment Group report a 2- and 5-year risk of At 2 years, no test showed any significant difference
severe neurotoxicity at 3% and 10%, respectively in from baseline.
patients receiving PCI (Shaw et al. 1994). In the UKCCCR/EORTC trial, 136 patients par-
A retrospective study of 30 patients performed at ticipated in extensive psychometric testing that was
MD Anderson Cancer Center attempted to better performed at randomization and every six months
characterize the risks of neurotoxicity from PCI (Gregor et al. 1997). Up to 41% of patients analyzed
518 M. C. Stauder and Y. I. Garces

had significant abnormalities on individual tests worsening in global health status (35 vs. 22%) from
before PCI and additional impairment was seen at base line up to 3 months. There was poor compliance
6 months and 1 year. There were no notable differ- with follow-up assessments with a 94% participation
ences, however, between the group receiving PCI and rate at baseline, but only 60 and 55% at 6 weeks and
the group that did not. The most common symptoms 3 months, respectively. Also, the median survival of
reported at follow-up were tiredness, lack of energy, 6 months seen in this cohort was shorter than
irritability, decreased sexual interest, shortness of expected. Both of these factors may have contributed
breath, and cough. These symptoms were as moderate to a lack of power to detect a difference between
or severe more frequently in the group of patients who groups. An exploratory analysis of other symptom
did not receive PCI. scale factors, however, showed significant differ-
Based on the increased local control in the brain ences, both statistical and clinical in appetite loss,
with the use of PCI in SCLC, recent trials have also constipation, nausea and vomiting, social functioning,
focused on patients with NSCLC. A prematurely future uncertainty, headaches, motor dysfunction, and
closed, prospective randomized trial enrolled 340 weakness of the legs favoring the control arm.
patients without disease progression after completing
definitive therapy for NSCLC and was randomized to
PCI (30 Gy/15 Fx) or observation (Sun et al. 2011).
Patients had neurocognitive function assessed with 5 Patterns of Care
Mini-Mental Status Exam (MMSE), Hopkins Verbal
Learning Test (HLVT), and Activity of Daily Living Using a complex decision-analytic model, and based
Scale (ADLS) and used the same quality of life tools on a large cohort of simulated patients, an assessment
as the patients described in the EORTC extensive of the potential overall value of PCI was explored
stage SCLC study (Slotman et al. 2007, 2009). No (Lee et al. 2006). Assumptions such as a low (30%)
significant differences in quality of life, MMSE, or and moderate (50%) latent rate of neurotoxicity and a
ADLS were observed at 1 year, but patients receiv- 5-year overall survival of a PCI and no-PCI group
ing PCI had decreased scores for immediate and derived from data published as part of a large Inter-
delayed recall (p = 0.03 and 0.008, respectively) on group SCLC trial (26 and 22%, respectively) were
HVLT. used in the model during patient simulation (Turrisi
et al. 1999). This resulted in a 7.8 and 13% incidence
of observed neurotoxicity at the low and medium
4.3 Extensive Stage Small-Cell Lung rates, respectively. This was then translated to a
Cancer quality-adjusted life expectancy (QALE) for each
group of patients to incorporate the impact of chronic
The effect of PCI on quality of life end points was neurotoxicity on patient quality of life. According to
also addressed in patients with extensive stage lung the model, in all cases when the latent neurotoxicity
cancer (Slotman et al. 2007, 2009). Two quality of rate is less than 54% or 5-year survival is less than
life tools (EORTC-QLQ-C30 and BN20) were used to 46%, the group of patients receiving PCI has a greater
analyze short and long-term changes in functioning. calculated QALE than those in the no-PCI group.
In a preliminary report, the authors note significant It is clear, however, that many patients who are
side-effects of PCI, including fatigue and hair loss, otherwise eligible to undertake PCI as part of their
which were significantly more severe in the group of treatment for SCLC, are not receiving it. A recently
patients receiving PCI (Slotman et al. 2007). No published study of 207 patients without progressive
significant differences were seen in global health disease after chemotherapy and thoracic radiotherapy
status, role functioning, cognitive functioning, or shows that only 61% actually completed a PCI regi-
emotional functioning. A subsequent study showed a men (Giuliani et al. 2010). Of these patients, 37.5% of
limited effect of PCI on these factors, but none patients refused PCI. In this cohort, over half (53%)
reached the level of clinical significance as pre- cited potential concerns about toxicity and another
defined in the protocol design (Slotman et al. 2009). 20% of patients cited excessive toxicity of their prior
Patients receiving PCI had an increased rate of severe chemoradiotherapy as a reason to refuse PCI.
Prophylactic Cranial Irradiation in Small-Cell Lung Cancer 519

part of comprehensive treatment for SCLC. Certainly,


6 Future Directions well-designed prospective clinical trials addressing
these issues are needed and are ongoing.
Recently, attempts have been made to decrease the
potential neuropsychological effects of whole brain
radiotherapy (WBRT). There is good evidence that
radiation disrupts the normal microvascular angiogenesis References
of the hippocampus and decreases neurogenic cell pro-
liferation in vitro (Monje et al. 2002). Abnormal hippo- Abayomi OK (1996) Pathogenesis of irradiation-induced cog-
campal function has been shown to correlate with nitive dysfunction. Acta Oncol 35(6):659–663
Arriagada R, Le Chevalier T et al (1995) Prophylactic cranial
decreased memory in both humans and animals irradiation for patients with small-cell lung cancer in
(Abayomi 1996; Broadbent et al. 2004). As such, several complete remission. J Natl Cancer Inst 87(3):183–190
groups have published their experience with hippocam- Aupérin A, Arriagada R et al (1999) Prophylactic cranial
pal avoidance in WBRT for brain metastases (Gutierrez irradiation for patients with small-cell lung cancer in
complete remission. N Engl J Med 341(7):476–484
et al. 2007; Gondi et al. 2010; Hsu et al. 2010). A Phase II Bremnes RM, Sundstrom S et al (2001) Multicenter phase II
clinical trial evaluating the effect of hippocampal trial of paclitaxel, cisplatin, and etoposide with concurrent
avoidance in WBRT is scheduled to begin enrollment radiation for limited-stage small-cell lung cancer. J Clin
soon and will evaluate delayed recall as assessed by the Oncol 19(15):3532–3538
Broadbent NJ, Squire LR et al (2004) Spatial memory,
HVTL at 4 months after hippocampal avoidance during recognition memory, and the hippocampus. Proc Natl Acad
WBRT in patients with brain metastasis (RTOG 0933, Sci USA 101(40):14515–14520
NCT01227954). Also, a Phase III cooperative group Cox JD, Petrovich Z et al (1978) Prophylactic cranial
trial is currently examining the role of the drug 3, 5- irradiation in patients with inoperable carcinoma of the
lung: preliminary report of a cooperative trial. Cancer
dimethyladamantan-1-amine (Memantine) in preventing 42(3):1135–1140
neurocognitive dysfunction due to WBRT (RTOG 0614, Giuliani M, Sun A et al (2010) Utilization of prophylactic
NCT00566852). These trials will provide important cranial irradiation in patients with limited stage small cell
additional information on the neuropsychological effects lung carcinoma. Cancer 116(24):5694–5699
Gondi V, Tolakanahalli R et al (2010) Hippocampal-sparing
of WBRT and suggest ways to minimize them. whole-brain radiotherapy: a ‘‘how-to’’ technique using heli-
cal tomotherapy and linear accelerator-based intensity-modu-
lated radiotherapy. Int J Radiat Oncol Biol Phys 78(4):
7 Conclusions 1244–1252
Gore EM, Bae K et al (2011) Phase III comparison of
prophylactic cranial irradiation versus observation in
Prophylactic cranial radiation should be recom- patients with locally advanced non–small-cell lung cancer:
mended for all limited stage SCLC patients in com- primary analysis of radiation therapy oncology group study
plete remission or those who exhibit a good partial rtog 0214. J Clin Oncol 29(3):272–278
Gregor A, Cull A et al (1997) Prophylactic cranial irradiation is
response to chemoradiotherapy in order to decrease indicated following complete response to induction therapy
brain metastasis and improve survival. In patients in small cell lung cancer: results of a multicentre random-
with ES-SCLC, PCI should be recommended for all ised trial. Eur J Cancer 33(11):1752–1758
patients with any response (partial or complete) to Grosshans DR, Meyers CA et al (2008) Neurocognitive
function in patients with small cell lung cancer. Cancer
chemotherapy. The optimal dose and fractionation 112(3):589–595
regimen is unknown, but a commonly accepted Gutierrez AN, Westerly DC et al (2007) Whole brain radio-
standard is 25 Gy in 10 fractions for LS-SCLC and therapy with hippocampal avoidance and simultaneously
20 Gy in 5 fractions for ES-SCLC patients. Good data integrated brain metastases boost: a planning study. Int J
Radiat Oncol Biol Phys 69(2):589–597
characterizing the neuropsychological effects of PCI Hansen HH (1973) Should initial treatment of small cell
are ongoing and no definitive conclusions can be carcinoma include systemic chemotherapy and brain irradi-
made regarding long-term toxicity. As the survival of ation? Cancer Chemother Rep 3, 4(2):239–241
patients with SCLC improves, the controversies Hsu F, Carolan H et al (2010) Whole brain radiotherapy with
hippocampal avoidance and simultaneous integrated boost
regarding the neurotoxic effect of PCI may become for 1–3 brain metastases: a feasibility study using volumet-
more defined. Also, patients need to be better ric modulated arc therapy. Int J Radiat Oncol Biol Phys
informed of the potential risks and benefits of PCI as 76(5):1480–1485
520 M. C. Stauder and Y. I. Garces

Jackson DV Jr, Richards F 2nd et al (1977) Prophylactic cranial changing pattern with lengthening survival. Cancer 44(5):
irradiation in small cell carcinoma of the lung. a randomized 1885–1893
study. JAMA 237(25):2730–2733 Ohonoshi T, Ueoka H et al (1993) Comparative study of
Jeremic B, Shibamoto Y et al (1999) Role of radiation therapy prophylactic cranial irradiation in patients with small cell
in the combined-modality treatment of patients with exten- lung cancer achieving a complete response: a long-term
sive disease small-cell lung cancer: a randomized study. follow-up result. Lung Cancer 10(1–2):47–54
J Clin Oncol 17(7):2092 Shaw E, Su J et al (1994) Prophylactic cranial irradiation in
Komaki R, Cox JD et al (1981) Risk of brain metastasis from complete responders with small-cell lung cancer: analysis of
small cell carcinoma of the lung related to length of survival the mayo clinic and north central cancer treatment group
and prophylactic irradiation. Cancer Treat Rep 65(9–10): data bases. J Clin Oncol 12(11):2327–2332
811–814 Slotman B, Faivre-Finn C et al (2007) Prophylactic cranial
Komaki R, Meyers CA et al (1995) Evaluation of cognitive irradiation in extensive small-cell lung cancer. N Engl J
function in patients with limited small cell lung cancer prior Med 357(7):664–672
to and shortly following prophylactic cranial irradiation. Int Slotman BJ, Mauer ME et al (2009) Prophylactic cranial
J Radiat Oncol Biol Phys 33(1):179–182 irradiation in extensive disease small-cell lung cancer:
Laplanche A, Monnet I et al (1998) Controlled clinical trial of short-term health-related quality of life and patient reported
prophylactic cranial irradiation for patients with small-cell lung symptoms—results of an international phase III randomized
cancer in complete remission. Lung Cancer 21(3):193–201 controlled trial by the eortc radiation oncology and lung
Le Pechoux C, Dunant A et al (2009) Standard-dose versus cancer groups. J Clin Oncol 27(1):78–84
higher-dose prophylactic cranial irradiation (PCI) in patients Sun A, Bae K et al (2011) Phase III trial of prophylactic cranial
with limited-stage small-cell lung cancer in complete remis- irradiation compared with observation in patients with
sion after chemotherapy and thoracic radiotherapy (PCI 99– locally advanced non–small-cell lung cancer: neurocogni-
01, EORTC 22003–08004, RTOG 0212, and IFCT 99–01): a tive and quality-of-life analysis. J Clin Oncol 29(3):279–
randomised clinical trial. Lancet Oncol 10(5):467–474 286
Lee JS, Umsawasdi T et al (1987) Timing of elective brain Suwinski MD, PDR, MSSP, Lee MD et al (1998) Dose–
irradiation: a critical factor for brain metastasis-free survival response relationship for prophylactic cranial irradiation in
in small cell lung cancer. Int J Radiat Oncol Biol Phys small cell lung cancer. Int J Radiat Oncol. Biol Phys
13(5):697–704 40(4):797–806
Lee JJ, Bekele BN et al (2006) Decision analysis for Tai P, Tonita J et al (2003) Twenty-year follow-up study of
prophylactic cranial irradiation for patients with small-cell long-term survival of limited-stage small-cell lung cancer
lung cancer. J Clin Oncol 24(22):3597–3603 and overview of prognostic and treatment factors. Int J
Levitan N, Dowlati A et al (2000) Multi-institutional phase I/II Radiat Oncol Biol Phys 56(3):626–633
trial of paclitaxel, cisplatin, and etoposide with concurrent Turrisi AT, Kim K et al (1999) Twice-daily compared with
radiation for limited-stage small-cell lung carcinoma. J Clin once-daily thoracic radiotherapy in limited small-cell lung
Oncol 18(5):1102–1109 cancer treated concurrently with cisplatin and etoposide.
Meert AP, Paesmans M et al (2001) Prophylactic cranial N Engl J Med 340(4):265–271
irradiation in small cell lung cancer: a systematic review of Wolfson AH, Bains Y et al (2001) Twice-daily prophylactic
the literature with meta-analysis. BMC Cancer 1:5 cranial irradiation for patients with limited disease
Monje ML, Mizumatsu S et al (2002) Irradiation induces neural small-cell lung cancer with complete response to
precursor-cell dysfunction. Nat Med 8(9):955–962 chemotherapy and consolidative radiotherapy: report of
Nugent JL, Bunn PA Jr et al (1979) CNS metastases in small a single institutional phase II trial. Am J Clin Oncol
cell bronchogenic carcinoma: increasing frequency and 24(3):290–295
Radiation Therapy for Lung Cancer
in Elderly
Branislav Jeremić and Željko Dobrić

Contents Abstract
Western world is rapidly aging, yet the exact threshold
1 Introduction.............................................................. 523 age for differentiating elderly versus non-elderly is not
2 Non-Small-Cell Lung Cancer................................. 525 widely adopted. Elderly are also underrepresented in
2.1 Early Stage Non-Small-Cell Lung Cancer ............... 525 clinical trials and level of evidence of preferred
2.2 Locally Advanced Non-Small-Cell Lung Cancer .... 529 treatment in both non-small-cell and small-cell lung
2.3 Metastatic Non-Small-Cell Lung Cancer ................. 532
cancer is basically lacking. From the existing evi-
3 Small-Cell Lung Cancer ......................................... 533 dence, however, it seems that radiation therapy is safe
4 Conclusions ............................................................... 538 and effective treatment modality in patients with early
stage non-small-cell lung cancer. It is so also in locally
References.......................................................................... 538
advanced non-small-cell lung cancer when combined
with radiochemotherapy, although caution should be
exercised when one attempts to combine high-dose
radiation therapy with high-dose chemotherapy. Sim-
ilarly, palliative radiation therapy is effective in
elderly with non-small-cell lung cancer unsuitable
for radical approach, with or without additional
chemotherapy. Finally, in small-cell lung cancer,
again, radiation therapy can successfully be employed
either alone or, most frequently, with chemotherapy in
treatment of both limited and extensive-disease small-
cell lung cancer. While largely unsubstantiated fear
has governed our approaches in elderly with lung
cancer in recent decades, novel radiation therapy
technologies and novel drugs should provide suitable
framework for embarking on more clinical trials
in elderly.

1 Introduction

Epidemiologic data are clearly showing that the


B. Jeremić (&)  Ž. Dobrić
Institute of Lung Diseases, Sremska Kamenica, Serbia Western world is rapidly ageing. Individuals over the
e-mail: nebareje@gmail.com age of 65 years compose the fastest-growing segment

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 523
DOI: 10.1007/174_2011_254, Ó Springer-Verlag Berlin Heidelberg 2011
524 B. Jeremić and Ž. Dobrić

of the population. In the year 1930, 1 out of 15 per- incidence is 69 years in males and 67 years in
sons was[65 years of age, while in the year 1990 this females. In patients with early/localized disease
was 1 out of 8. It is estimated that by the year 2020, treatment approaches with curative intention are fea-
they will constitute approximately 20% of all popu- sible. However, the widely accepted ‘‘evidence’’ is
lation in the United States, and by the year 2030 usually based on studies which are performed with
they will constitute approximately 25% of all pop- selected patients, the elderly patients being under-
ulation in the United States. The group [75 years represented in clinical trials. Disturbing observation
old will triple by 2030, and those [85 years of age from daily practice worldwide is that elderly are less
will double in the same period (Yanick 1997). likely to be vigorously screened and staged, and fre-
Unfortunately, no widely-accepted and exact defini- quently less aggressively treated (Nugent et al. 1997;
tion of an elderly person exists. Cut-off age thresh- Higton et al. 2010).
olds differentiating an elderly versus non-elderly Importantly, though, when evaluated for the spe-
person vary between 60 and 80 years of age, many cific features they did not seem to have different
studies using the cut-offs between 65 and 75 years. characteristics at presentation. This particularly
Contrasting fixed thresholds are used to define elderly relates to the stage of the disease, performance status,
persons, operational definition of geriatric oncology is and histology, when compared to their non-elderly
‘‘when the health status of a patient population begins counterparts, although other characteristics such as
to interfere with the oncological decision-making type and number of co-morbidities and organ function
guidelines’’ (Extermann 2000). Hence, rather than an differ in the two groups of populations (Montella et al.
arbitrarily established age limit, biological age should 2002). Furthermore, there still may be a reduction in
be defined individually by performance status and functional organ capacity although not clinically overt
co-morbidities, the two ultimately influencing the and undetected in many ‘‘healthy’’ elderly. With
decision–making process. increasing age kidney, lung, and heart as well as the
Increasing age is direct consequence of improved immune system may show a reduced organ function
sanitation, use of antibiotics, reduced smoking, and (Balducci and Extermann 2000a).
diet. It has, however, many consequences, among To base patient selection on clinical judgement with
which there is a particular one, directly-associated performance status and organ function parameters is a
increasing cancer occurrence rates: eleven fold common practice in oncology. When one consider
increase in the cancer incidence in persons [65 years elderly this, however, may not be adequate, since there
of age when compared to their younger counterparts is a need for a more comprehensive pre-treatment
indicates that elderly population will soon become assessment that would consider potential hazards in
one of the major targets in oncology requiring specific treating elderly the same way as non-elderly. This may
managements for various cancers (Yanick 1997). help in predicting and avoiding such hazards
Indeed, when compared to the general population of (Monfardini et al. 1995), since geriatric syndromes
the year 1990, in the year 2010 it is estimated that widely recognized as potential threat to diagnosis and
there will be a growth of 9% in the general, but treatment include dementia, depression, incontinence,
growth in cancer cases will be more than 30%. In the delirium, osteoporosis, and failure to thrive. A com-
year 2020, it is estimated that population growth prehensive geriatric assessment as an adjunct to general
would be 12% while growth of cancer population and cancer-specific diagnostic procedures is therefore,
would be 60%. There are multiple reasons for such developed. It has been defined as a multidimensional,
growth in cancer incidence such as less resistance and interdisciplinary diagnostic process to determine the
longer exposure to carcinogens, reduced immune medical, psychological, and functional capabilities of a
competence, altered anti-tumor defence, decreased frail elderly person in order to develop a coordinated
DNA repair, defects in tumor suppressor genes, and and integrated plan for treatment and long-term follow-
biologic behavior differences. up (Rubenstein 1995; Osterweil et al. 2000; Bernabei
Lung cancer, major cancer killer in both sexes, is a et al. 2000). This assessment includes the medical
typical disease of the elderly patient. More than 50% assessment, assessment of functioning, psychological
of patients are over the age of 65 years and nearly assessment, social assessment, and environmental
35% are over the age of 70 years. The median age of assessment (Balducci and Extermann 2000b).
Radiation Therapy for Lung Cancer in Elderly 525

Finally, to set up a stage and scene for further cancer, with or without additional chemotherapy,
discussions, one may start with recent analysis whereas the standard treatment for locally advanced
(Owonikoko et al. 2007) which studied the burden non-small-cell lung cancer is not well-defined.
and outcome of lung cancer in elderly by focusing on Although a minority of Stage IIIA non-small-cell lung
cancer outcomes in the period 1988–2003 from cancer patients can be treated with surgery alone,
Surveillance, Epidemiology, and End Results data- there is no clear benefit for either surgery alone or
base. Study subjects were split into those C80 years surgery preceded by radiation therapy and chemo-
old versus those 70–79 years old versus those therapy. In addition, the vast majority of patients with
\70 years old. Data used included demographics, Stage III non-small-cell lung cancer are unresectable,
stage, and 5-year relative survival, the latter one and, therefore, best treated with concurrent radiation
investigated upon influence of histology, sex, race, therapy and chemotherapy.
stage and treatment. Two temporal trends (1988–1997
and 1997–2003, respectively) were used. Distribution
by stage and histology was comparable for three 2.1 Early Stage Non-Small-Cell Lung
groups. Five year overall survival was lower in very Cancer
elderly (7.4 versus 12.3 versus 15.5%) (P \ 0.0001)
across sex, histology subtype, stage, and race. Patients Nearly 25% of elderly with early stage non-small-cell
C80 years were less likely to receive local treatment lung cancer do not undergo surgery (Bach et al.
(no surgery or radiation therapy) than younger 1999). Among the reasons for not undergoing surgery,
patients: 47 versus 28 versus 19% (C80 versus 70–79 besides their advanced age, elderly were frequently
versus \70 years). Overall outcome for patients discriminated due to existing co-morbidity and,
receiving surgery or radiation therapy was compara- only occasionally, there was a refusal to undergo
ble across the three age groups, but elderly had a surgery. In the last decade, two studies addressed the
poorer outcome when radiation therapy or surgery issue of the survival of unresected patients treated
were combined or when no therapy was instituted. with and without radiation therapy, showing that
This chapter addresses some of the important issues radiation therapy may improve survival of these
in radiation therapy of lung cancer in elderly. Widely patients. In the first study, Wisnivesky et al. (2005),
accepted clinical designation of non-small-cell lung however, used only a standard regression to correct
cancer and small-cell lung cancer as two separate for potential selection bias and the analyses were not
entities will here serve as well to enable suitable adjusted for use of chemotherapy or co-morbidities.
framework for addressing these issues in elderly. As patients with higher number of co-morbidities
were less likely to receive radiation therapy and/or
chemotherapy, lack of adjustments of these factors
2 Non-Small-Cell Lung Cancer may have biased the analyses toward showing a sig-
nificant effect of radiation therapy on survival. These
Non-small-cell lung cancer accounts for approxi- observations may have prompted same group of
mately 80% of all lung cancer cases, with[50% of all investigators (Wisnivesky et al. 2010) to undertake
patients with non-small-cell lung cancer being similar analysis using now a propensity score and
[65 years old with about one-third of all patients instrumental variable analyses to minimize potential
being[70 years old at the time of diagnosis. Curative biases. Authors have used Surveillance, Epidemiol-
approaches are feasible in patients with early Stage ogy, and End Results data identifying a total of
(I–II) disease and in a proportion of patients with [6,000 eligible patients. Overall, 59% patients
locally advanced disease (Stage IIIA/IIIB), while received radiation therapy. The overall and lung
palliation is the goal for the remainder of locally cancer-specific survival of unresected patients treated
advanced and all metastatic (Stage IV) non-small-cell with radiation therapy was significantly better com-
lung cancer patients. This general concept should also pared with the untreated cases (P \ 0.0001 for both
prove to be feasible in elderly with non-small-cell comparisons). Both propensity score and instrumental
lung cancer. Surgery is the treatment of choice for variable indicted improved outcomes among patients
patients in early Stage (I/II) non-small-cell lung treated with radiation therapy.
526 B. Jeremić and Ž. Dobrić

To further extend this issue, Palma et al. (2010a) survival, disease-specific survival or local progres-
used prospective databases from British Columbia to sion-free survival in elderly versus non-elderly.
identify patients with Stage I non-small-cell lung Wurschmidt et al. (1994) used multivariate analyses
cancer treated curatively with either surgery or radi- as well as did Kaskowitz et al. (1993) to show no
ation therapy between 2000 and 2006. Kaplan–Meier, difference in the treatment outcome between elderly
Cox regression, and competing-risk analyses were and non-elderly. The same observation was made by
used to assess overall survival and disease-specific Slotman et al. (1994), Gauden et al. (1995), and Krol
survival in the elderly, and the relationship between et al. (1996). In the two of studies of Jeremic et al.
age and survival outcomes. Of a total of 558 patients (1997, 1999a) no difference in either survival or
with Stage I disease, 310 (56%) received surgery, and relapse-free survival was observed between patients
248 (44%) received radiation therapy. Elderly patients \60 years old and those C60 years old with Stage I
(age C 75 years) were less likely to undergo resection and II non-small-cell lung cancer, respectively, trea-
than their younger counterparts (43 versus 72%, ted with hyperfractionated radiation therapy alone
P \ 0.0001). Actuarial overall survival after surgery with a total dose of 69.6 Gy using 1.2 Gy b.i.d.
for elderly patients was 87% at 2 years and 69% at fractionation. Multi-variate analyses using both sur-
5 years. On multivariate analysis, overall survival vival and relapse-free survival confirmed that age did
after surgery was dependent on tumor stage (P = not play an important role in this setting.
0.034) and performance status (P = 0.03), but not age In a study of Hayakawa et al. (1999) 97 patients
(P = 0.87). After radiation therapy, actuarial overall C75 years old (elderly) and 206 patients \75 old
survival for elderly patients was 53% at 2 years and (non-elderly) were treated with radiation therapy
23% at 5 years. On multivariate analysis, age did not doses C60 Gy (up to [80 Gy) for inoperable non-
predict for overall survival after radiation therapy small-cell lung cancer. Elderly were classified into the
(P = 0.43), whereas tumor stage (P = 0.033), sex two subgroups: A—75–79 years and B—C80 years.
(P = 0.044), and dose (P = 0.01) were significant No difference was found between the three age groups
predictors. This study reconfirmed earlier observa- (5-year survival: 12 versus 13 versus 4% for non-
tions that survival after radical treatment for Stage I elderly, elderly A, and elderly B, respectively), but a
non-small-cell lung cancer is dependent on factors multivariate analysis disclosed detrimental effect of
such as tumor stage, performance status, sex, and oldest age, due to 14% treatment-related deaths in
radiation therapy dose, but not age. This has again patients receiving 80 Gy. Unfortunately, no multi-
indicated that elderly patients who are sufficiently fit variate analysis was done using disease-specific sur-
should not be considered ineligible for radical treat- vival as endpoint to give better insight into this
ment based on age alone. finding and help to identify predisposing factors
Data from the existing literature, accumulated over leading to more treatment-related deaths in elderly
several decades, clearly shows that radiation therapy group treated with high-dose radiation therapy.
is effective in this setting. Aristizabal et al. (1976) Gauden and Tripcony (2001) investigated the
seems to be very first to show that elderly (C70 years) effect of age using either dividing study group into the
had significantly better 2- year survival than 5-year subgroups or using a specified cut-off value
non-elderly (49–69 years) (35.7 versus 13.1%, P = (\70 years versus C70 years) on treatment outcome
0.044), likely as a consequence of high local control in patients with Stage I non-small-cell lung cancer.
(70%) and a lower incidence of distant metastasis. The median survival times (22 versus 26 months) and
Similarly, Coy and Kennelly (1980) and Newaishy a 5-year survival (22 versus 34%), respectively for
and Kerr (1989) observed a significant tend toward non-elderly and elderly were observed. The same held
better survival in older patients with non-small-cell true for recurrence-free survival. Finally, both overall
lung cancer treated with radiation therapy alone. In survival and recurrence-free survival remained simi-
addition, Noordijk et al. (1988) found no difference in lar in various 5-year subgroups. The multivariate
the outcome of elderly patients treated with radiation analysis excluded age as an important prognostic
therapy alone or surgery. In radiation therapy alone factor predicting either of these two endpoints.
studies, Sandler et al. (1990) and Rosenthal et al. In contrast, Morita et al. (1997) found a survival
(1992) found no significant difference in overall advantage for patients \80 years old when compared
Radiation Therapy for Lung Cancer in Elderly 527

to those C80 years old (5-year survival rate: 25.2 therapy-induced acute lung toxicity, while only two
versus 7.7%), but did not specify the outcome when (6%) patients experienced late grade 3 lung toxicity.
splitting \80 year old group into various subgroups No other high-grade toxicity was observed during this
(e.g. \70 versus 70–80 years). Similarly, Sibley et al. study. Ahmad et al. (2010) treated patients aged
(1998) documented superior outcome in younger 60 years and older with inoperable Stage I non-small-
(\60 years) patients with Stage I versus older cell lung cancer using a hypofractionated schedule.
patients, unconfirmed, however, when local progres- Between 1991 and 2006, 75 such patients were
sion was used as an endpoint (P = 0.10). identified with median age of 74 years (range,
While some studies attempted to evaluate toxicity, 60–86). Patients received a median total dose of
in none of these series it was specified that these toxic 65 Gy using median daily dose fractions of 2.5 Gy.
events have happened in elderly. In those studies Median local failure free survival was 19.6 months
which specifically addressed elderly with early stage and median overall survival was 21.2 months. Anal-
non-small-cell lung cancer, no significant radiation ysis of competing risks showed that at 5 years, the
therapy-related complications were found and inci- probability of local failure as the first detected event
dence of both acute and late high-grade toxicity was was 22.1%, the probability of distal failure as the first
similar among all age groups (Gauden and Tripcony detected event was 14.5%, and the probability of
2001). When radiation therapy-related deaths occurred, death without recording a failure was 48.6% Radia-
again, there was no difference between elderly (5%) tion-related toxicity of grade 3 or greater was seen in
treated with highest dose levels (80 Gy) and their three patients and there were no treatment-related
non-elderly counterparts (4%) treated the same way deaths.
(Hayakawa et al. 2001). Confirmation of the importance of novel radiation
When these data are summarized, conventionally therapy technologies comes from the study of Park
planned external beam radiation therapy appears et al. (2010) who investigated whether complex
capable of producing the median survival times of radiation therapy planning was associated with
20–27 months and 5-year survivals of 15–34% in improved outcomes in a cohort of elderly patients
patients [70 years and event better results when the with unresected Stage I–II non-small-cell lung cancer.
cut-off of 60 years is used. Using the Surveillance, Epidemiology, and End
Results of studies published in recent years seem to Results registry linked to Medicare claims, they
confirm these observations. In a study by San Jose identified 1998 patients aged [65 years with histo-
et al. (2005), covering a period between 1995 and logically confirmed, unresected Stage I–II non-small-
1999, a total of 33 male patients were treated with cell lung cancer. Patients were classified into an
radiation therapy alone. Radiation therapy doses intermediate or complex radiation therapy planning
ranged 66–78 Gy (median, 70 Gy) using standard group using Medicare physician codes. To address
fractionation (2.0 Gy per fraction). The age range was potential selection bias, they used propensity score
71–97 years (median, 75 years) with 11 patients modelling. Survival of patients who received inter-
being C80 years old. Twenty-two (67%) patients had mediate and complex simulation was compared using
a squamous cell carcinoma. There were 24 (73%) Cox regression models adjusting for propensity scores
Stage I and nine (27%) Stage II patients. Radio- and in a stratified and matched analysis according to
graphic objective response rate was observed in 23 propensity scores. Overall, 25% of patients received
(70%) patients. The median survival time was complex radiation therapy planning. Complex radia-
37.4 months and 3-year survival time was 50%, while tion therapy planning was associated with better
the median cause-specific survival time was overall (hazard ratio 0.84; 95% confidence interval,
48.1 months and a 3-year cause-specific survival rate 0.75–0.95) and lung cancer-specific (hazard ratio
was 55.3%. The median time to local recurrence was 0.81; 95% confidence interval, 0.71–0.93) survival
36.8 months and a 3-year local recurrence-free sur- after controlling for propensity scores. Similarly,
vival rate was 50.2%, while the median time to distant stratified and matched analyses showed better overall
metastasis was not achieved yet at the time of that and lung cancer-specific survival of patients treated
report, the 3-year distant metastasis-free survival rate with complex radiation therapy planning. The use of
being 71.4%. One (3%) patient died of radiation complex radiation therapy planning in this study was
528 B. Jeremić and Ž. Dobrić

associated with improved survival among elderly 299 patients (34%), radiation therapy in 299 patients
patients with unresected Stage I–II non-small-cell (34%), and neither in 277 patients (32%). Radiation
lung cancer. These findings should be validated in therapy use increased between periods A and C (26
prospective randomized controlled trials. versus 42%, P \ 0.01), corresponding to a decrease
Recent years also brought clinical data from in untreated patients. The percentage of radiation
studies which used sophisticated treatment planning therapy patients undergoing stereotactic radiation
and delivery in this population. In the study of Niibe therapy in periods B and C was 23 and 55%,
et al. (2003), 22 elderly with tumors up to 5 cm were respectively. Median survival for all patients
treated with fraction size of 3–4 Gy, five fractions per increased from 16 months in period A to 21 months
week with a mean total dose of 65.3 Gy. Overall in period C (log-rank P \ 0.01). The improvement in
survival rates at 1–3 years were 100, 83, and 56%, overall survival was confined to radiation therapy
respectively, while local control rates at 1–3 years patients, whereas no significant survival improve-
were 92, 83, and 83%, respectively. There was no ments were seen in the other groups. Stereotactic
grade 2 or greater toxicity except in two patients. radiation therapy introduction was associated with a
These results showed that this tool, now widely 16% absolute increase in radiation therapy use, a
available worldwide, provide feasible and effective decline in the proportion of untreated elderly patients,
radiation therapy treatment in elderly. Yu et al. (2008) and an improvement in overall survival. Indeed, in the
investigated the efficacy and safety of involved-field study of Haasbeek et al. (2010), 203 tumors in 193
radiotherapy in 80 patients who are 70 years old or patients aged C75 years were treated using stereo-
more with early Stage (I/II) non-small-cell lung tactic radiation therapy. The median patient age was
cancer using intensity modulated radiation therapy. 79 years, 80% of patients were considered medically
Intensity modulated radiation therapy plans were inoperable, and 20% of patients declined surgery.
designed to deliver 66.6 Gy to involved-field. The Risk-adapted stereotactic radiation therapy regimens
objective response rate of all patients was 88.6% with were used with the same total dose of 60 Gy in 3
a median overall survival time of 38 months and the fractions (33%), 5 fractions (50%), or 8 fractions
1-, 2- and 5-year overall survival rates and local (17% of patients), depending on the patient’s risk for
progression-free survival rates were 65.8, 55.7, 25.3, toxicity. All but one patient completed treatment.
and 84.8, 59.5, and 34.2%, respectively. Twenty-nine Survival rates at 1 and 3 years were 86 and 45%,
patients (36.7%) with elective nodal failures were respectively. The actuarial local control rate at
identified, with a median time to treatment failure of 3 years was 89%. Acute toxicity was uncommon and
55 months (range 49–61 months) after treatment. late Radiation Therapy Oncology Group grade C3
There were no treatment-related deaths or grade 4 toxicity was observed in\10% of patients. This study
toxicity. Grade 3 toxicities were esophagitis (1.3%), showed that, taking into account risks and existing co-
radiation pneumonitis (3.8%), and hematological morbidities bring, elderly could achieve the same
effects (2.5%). outcome as their non-elderly counterparts when
Stereotactic radiation therapy also found a fruitful treated with the same technique.
soil in elderly with early, mostly Stage I non-small- Most recently, however, the very first study com-
cell lung cancer. In a study from The Netherlands pared surgery with continuous hyperfractionated
(Palma et al. 2010b), the impact of introducing ste- accelerated radiation therapy in elderly with Stage I
reotactic radiation therapy in patients 75 years of age non-small-cell lung cancer (Ghosh et al.2003). One-
or older was studied using a population-based cancer hundred forty-nine patients underwent lobectomy, 47
registry. Amsterdam Cancer Registry was assessed in had wedge resection, while 19 had radiation therapy
three eras: 1999–2001 (period A, pre-stereotactic alone. Non-lobectomy patients had significantly lower
radiation therapy), 2002–2004 (period B, some pulmonary function. Survival at 1 and 5 years was 97
availability of stereotactic radiation therapy), and and 68% versus 98 and 74% versus 80 and 39%,
2005–2007 (period C, full access to stereotactic respectively (P = 0.0484). While survival data
radiation therapy. A total of 875 elderly patients were favored surgical approaches, this was associated with
diagnosed with Stage I non-small-cell lung cancer in a 2.7% 30 day operative mortality in the lobectomy
the study period. Primary treatment was surgery in group. Importantly, the frequency of loco-regional
Radiation Therapy for Lung Cancer in Elderly 529

recurrence was similar between the groups, while no Organization for Research and Treatment of Cancer
other endpoints were used in that study (e.g. cause- (Pignon et al. 1998). When adjusted for the primary
specific survival). This study showed again that location of the tumor, survival was comparable in
radiation therapy alone is a reasonable treatment each group. Distribution over age was similar for
option for those who are not suitable candidates for acute nausea, dyspnoea, esophagitis, weakness, and
surgery. the World Health Organization performance status
alteration. Also, the minimal time for complication
was similar in all age groups. No difference between
2.2 Locally Advanced Non-Small-Cell age groups was found regarding the patients experi-
Lung Cancer encing no complications at 4 years post-treatment.
Only grade 2 late esophagitis demonstrated a signifi-
Some of the studies which included ‘‘inoperable cant trend to be more frequent in older patients
cases’’ also included a proportion of patients with (P = 0.01), but this difference disappeared after
Stage III; other did not specify outcome regarding adjustment on study (P = 0.32). Gava (1999) repor-
age. Due to poor results of early studies, some ted on Italian Geriatric Radiation Oncology Group
suggested prohibition of radiation therapy in patients study on outcome of radiation therapy alone in 38
over 70 years (Aristizabal et al. 1976; Patterson et al. elderly patients with Stage III non-small-cell lung
1998). In a retrospective study of Nakano et al. (1999) cancer in whom 1-year survival rate approached 44%.
elderly patients with Stage III non-small-cell lung Another Italian study provided data which confirmed
cancer who had been treated with radiation therapy effectiveness of radiation therapy in 48 patients
alone were investigated. It resulted in a median sur- C75 years with locally advanced non-small-cell lung
vival time of 11.5 months in the younger group and cancer (Lonardi et al. 2000). Radiation therapy alone
6.3 months in the elderly group (P = 0.0043). In was used to give a median dose of 50 Gy. Overall
multivariate analysis not only a good performance survival was 10% at 24 months. There seems to be a
status, and good response but also age less than dose-response observed in that study with elderly
75 years as well were significantly favorable and treated with C50 Gy achieving a significantly better
independent predictors of survival. There were more survival than those treated with \50 Gy (the median
deaths due to respiratory infections and lower prog- survival time, 8 versus 4 months; 2-year survival, 20
nostic nutritional indexes before and after radiation versus 4%, respectively; P = 0.03). Tombolini et al.
therapy in elderly group patients. Hayakawa et al. (2000) also analysed patients C70 years in Stage III
(2001) also reported on an inferior survival in sub- non-small-cell lung cancer treated with radiation
group of elderly patients with Stage III disease. therapy alone using doses of 50–60 Gy (and a 10 Gy
However, this was noted only in patients[80 years of boost to the gross tumor volume) in 1.8–2 Gy frac-
age. Contrary to that, Kusumoto et al. (1986) inves- tions. Two-year overall and disease-free survival was
tigated this effect in Stage III/IV non-small-cell lung 27 and 14.6%, respectively. More recently, Pergolizzi
cancer where patients \70 years old (n = 64) and et al. (2002) reported on radiation therapy alone in 40
those C70 years old (n = 36) achieved the median elderly patients with Stage IIIA using a median dose
survival time of 7 and 6 months, respectively, the of 60 Gy, conventionally fractionated. No treatment-
difference being insignificant. related mortality was observed and no clinically sig-
Many studies, however, provided data on the nificant acute morbidity was scored. The median
effectiveness of radiation therapy in elderly. Zachariah survival time was 19 months and 5-year survival was
et al. (1997) reported on radiation therapy with 12%.
59–66 Gy using standard fractionation used in lung For many years now, radiochemotherapy is a
cancer in octogenarians. Response was observed in widely used approach in patients with locally
43% patients, and only 24% had progressive disease. advanced non-resectable non-small-cell lung cancer
Another study evaluated influence of the age on and good performance status. One may, therefore,
treatment outcome as well as acute and late toxicity of rightly wonder whether this is also true for elderly
curative thoracic radiotherapy using the data of 1,208 with the same disease. Some of the available studies
patients enrolled in trials conducted by the European provided retrospective subgroup (age) analyses of
530 B. Jeremić and Ž. Dobrić

patients enrolled into radiochemotherapy trials but treatment outcome. Patients [70 years (n = 104)
could not identify age as negative prognostic factor in were compared to those younger \70 years
multivariate analyses (Schaake-Koning et al. 1992; (n = 491). It was shown that elderly patients benefit
Jeremic et al. 1998a; Clamon et al. 1999; Furuse et al. from concurrent, as compared to sequential radio-
1999). Contrasting these results were the results of the chemotherapy in a similar magnitude as their younger
Radiation Therapy Oncology Group who reported on counterparts. Similar to the study of Rocha Lima et al.
a study which included 1999 patients treated (2002), the observation was that elderly people have
with radiation therapy with or without chemotherapy suffered from an increase in toxicity, especially severe
in several prospective studies. Using a recursive esophagitis. Recently, Schild et al. (2003) performed
partitioning and amalgamation analysis they have a secondary analysis of the North Central Cancer
found a negative influence of older age on survival Treatment Group study which evaluated split-course
(Werner-Wasik et al. 2000). These results recon- versus standard-fraction radiotherapy and cisplatin/
firmed their earlier results (Movsas et al. 1999) where etoposide in Stage III non-small-cell lung cancer. The
a quality-adjusted survival was used to examine 979 two age groups (\70 versus C70 years) achieved
patients with inoperable Stage II–IIIB non-small-cell similar 2- and 5-year survival rates, but grade C4
lung cancer patients treated with radiation therapy toxicity occurred in 62% patients \70 years of age
with or without chemotherapy enrolled into six pro- versus 81% in those C70 years of age (P = 0.007).
spective trials. Elderly patients had the best quality- Both grade C4 hematologic toxicity and grade C4
adjusted survival with radiation therapy alone, which pneumonitis were significantly more frequent in the
was in sharp contrast to their younger counterparts elderly group. More recently, another retrospective
who benefited mostly from more aggressive, com- North Central Cancer Group study (Schild et al. 2007)
bined radiochemotherapy approaches. The third ret- included the data from two prospective randomized
rospective analysis comprised of pooled data from trials comparing radiation therapy alone with com-
three trials at the same group (Langer et al. 2000) bined radiochemotherapy. In patients [65 years of
which showed that patients [70 years did not benefit age, combined radiochemotherapy was found to be
from concurrent radiochemotherapy when compared superior to radiation therapy alone. This was, how-
to those treated with radiation therapy alone. In spite ever, achieved at the expense of higher incidence of
of the fact that these three large analyses unequivo- toxicity in the combined-modality group. In a sub-
cally stand unified against the more intensive treat- group (age) analysis of the original Hoosier Oncology
ment approach in elderly, it must, however, be clearly Group study which showed the failure of use of
emphasized that they are difficult to interpret because consolidation chemotherapy in locally advanced non-
the compilation of patients treated on separate study small-cell lung cancer (Hanna et al. 2008, Sgroi et al.
protocols implies a comparison between patients with 2007) preliminarily reported that elderly had similar
a variety of entry criteria used to define eligibility and median survival time when compared to their non-
different treatment regimens administered, including a elderly counterparts (17.2 versus 21.2 months,
single-modality radiation therapy in many of these P = 0.3255). There was, however, higher incidence
studies. Contrary to these, Rocha Lima et al. (2002) of esophagitis (23 versus 15%) and dehydration (15
analyzed older patients from a randomized Cancer versus 7%) in elderly. In addition, elderly patients
and Leukemia group B trial of induction chemother- were more likely to discontinue treatment due to
apy followed by either radiation therapy alone or toxicity (12 versus 2%).
concurrent radiochemotherapy for locally advanced Some authors provided the data on prospective
non-small-cell lung cancer. Patients [70 years com- approaches addressing this issue. In one such attempt
pleted treatment to the same extent as younger between January, 1988 and June, 1993, Jeremic et al.
patients and attained similar response and survival, (1999b) enrolled a total of 58 patients into a phase II
but at the expense of increased toxicity, especially study, of whom 55 were evaluable. Carboplatin
high-grade ([3) nephrotoxicity and neutropenia. (400 mg/m2) was given intravenously on days 1 and
A retrospective analysis of the data from the Radia- 29, and etoposide (50 mg/m2) was given orally on
tion Therapy Oncology Group 94–10 study by Langer days 1–21 and 29–42. Accelerated hyperfractionated
et al. (2001) investigated the influence of age on radiation therapy was administered starting on day 1,
Radiation Therapy for Lung Cancer in Elderly 531

with a total dose of 51 Gy in 34 fractions over control, median survival time and 5-year survival were
3.5 weeks. A complete response rate was 27% and the all significantly improved in the cisplatin group (81%,
overall response rate was 65%. The median time to 33.4 months, and 38.3 versus 38.1%, 9.8 months, and
local recurrence was 14 months and the 5-year local 4.2%, respectively; local control, P \ 0.01; survival,
control rate was 13%, while the median time to dis- P \ 0.001). Multivariate analysis showed that addition
tant metastasis was 18 months and the 5-year distant of cisplatin via supra-selective way (bronchial artery
metastasis-free rate was 15%. The median time until infusion) was the strongest predictor of improved sur-
relapse was 8 months and the 1-, 2-, and 5-year vival achieved at no increase in life-threatening toxic-
relapse-free survival rates were 45, 20, and 9.1%, ity. Most recently, a phase II study (Giorgo et al. 2007)
respectively. The median survival time was evaluated safety and efficacy of sequential platinum-
10 months, and the 1-, 2-, and 5-year survival rates based chemotherapy followed by concurrent radio-
were 45, 24, and 9.1%, respectively. Hematological, chemotherapy in 30 consecutive patients with locally
esophageal, and bronchopulmonary acute grade 3/4 advanced non-small-cell lung cancer. Age ranged
toxicities were observed in 22, 7, and 4% of the 70–76 years (mean, 73 years). The median survival
patients, respectively. Neither grade 5 toxicity nor late time was 15 months and the median progression-free
grade [3 toxicity was observed. The survival results survival was 8.7 months. Grade 3–4 neutropenia
appeared to be comparable to those obtained in non- developed in 12 (40%) patients, while two patients
elderly patients with Stage III non-small-cell lung developed neutropenic fever. Grade 3 anemia and
cancer treated by full-dose radiation. In another phase grade 3 thrombocytopenia developed in three patients
II study, Atagi et al. (2000) used standard fraction each. Grade 3 esophagitis developed in six (20%)
radiation therapy with 50–60 Gy and concurrent low- patients during the concurrent radiochemotherapy part
dose daily carboplatin (30 mg/m2) to treat 38 patients of the treatment. No grade 5 toxicity was observed
with locally advanced non-small-cell lung cancer, 26 during this study. In the first prospective randomized
of whom were Stage III. The median survival time trial comparing radiation therapy alone (60 Gy in 30
was 15.1 months and 2-year survival was 20.5%. daily fractions) with the same radiation therapy and
Nakano et al. (2003) also reported on a pilot study in concurrent carboplatin given days 1–20 of the radiation
which low-dose cisplatin (6 mg/m2; days 1–5, 8–12, therapy course, Atagi et al. (2005) observed better
29–33, and 36–40) was added to conventionally median survival time (554 versus 428 days) and 1-year
fractionated radical radiation therapy (60 Gy given survival (65 versus 61%) for combined-modality
via 2.0 Gy daily fractions) in elderly with locally approach. However, this difference did not reach sta-
advanced unresectable non-small-cell lung cancer. Of tistical significance. Four patients died of treatment-
12 registered patients 11 were eligible for this anal- related toxicity (three in radiochemotherapy arm), 60%
ysis, 91% of whom were Stage III. The overall patients had protocol violations, 7% patients had vio-
response rate was 82%, the median overall survival lation in dose constraint to lung (two died of radiation
was 23 months and the 2-year survival rate was 53%, pneumonitis). This trial stopped early due to poor
respectively. The most common grade 3 toxicities accrual and, therefore, effectiveness of concurrent
included grade 3 leukopenia and thrombocytopenia radiochemotherapy approach as used in this study
occurring in 20 and 9%, respectively. No other high- remained unclear. Most recently, Jatoi et al. (2010)
grade toxicity was observed during this study. In an reported on a phase II study of cetuximab and radiation
attempt to selectively target tumor cells with cisplatin therapy in elderly and/or poor performance status
while decreasing the toxicity concurrent radioche- patients with locally advanced non-small-cell lung
motherapy may lead to, Karasawa et al. (2002) used cancer. Older patients [C65 years with an Eastern
bronchial arterial infusion of cisplatin and concurrent Cooperative Oncology Group performance status of 0,
radiation therapy (dose 50.4–73.2 Gy; median, 1, or 2] or younger patients (performance status of 2)
60.8 Gy) in 31 elderly Stage III non-small-cell lung received cetuximab 400 mg/m2 i.v. on day 1 followed
cancer. The results obtained that way were compared by weekly cetuximab 250 mg/m2 i.v. with concomitant
to that obtained in 30 elderly patients receiving no radiation of 60 Gy in 30 fractions. Patient median age
cisplatin. Response rate in the cisplatin group was 90 (range) was 77 years (60–87), and 12 (21%) had a
and 83% in the non-cisplatin group. Two-year local performance status of 2. The median survival was
532 B. Jeremić and Ž. Dobrić

15.1 months [95% confidence interval (CI) with non-surgical bimodality therapy or tri-modality
13.1–19.3 months], and the median time to cancer therapy including surgery had similar rates of grade 3/4
progression was 7.2 months (95% CI 5.8-8.6 months). toxicity (P = 0.18) and toxic death (P = 0.76). Sur-
No treatment-related deaths occurred, but 31 patients vival was worse with increasing age (P \ 0.0001),
experienced grade 3+ adverse events, most commonly likely due to greater use of palliative treatment in the
fatigue, anorexia, dyspnea, rash, and dysphagia, each of elderly. There was no difference between age groups
which occurred in\10% of patients. for non-surgical (P = 0.32) or surgical (P = 0.53)
In most of the studies the toxicity of the combined therapy when survival was analyzed for patients treated
treatment was tolerable (Jeremic et al. 1999b; Lonardi with curative intent. This study again reconfirmed
et al. 2000; Jatoi et al. 2010), with both acute and late previous observations that in select fit elderly patients,
high-grade toxicity not different from that observed combined-modality therapy is tolerable and is associ-
with similar approaches in non-elderly. Contrary to ated with survival similar to that of younger patients.
that, occasional study (Atagi et al. 2000) observed
high-grade hematological toxicity in 34.2–71.1% of
patients. Non-hematological toxicity was mild, with 2.3 Metastatic Non-Small-Cell Lung
no patient developing grade [3 esophagitis, although Cancer
two (5%) grade 4 pulmonary toxicities occurred.
It was also shown that co-morbidity and/or poor In spite of the fact that it is frequently used to treat
performance status and/or pronounced weight loss, intrathoracic or distant spread in Stage IV (metastatic)
rather than age, can influence patient selection for a non-small-cell lung cancer, there is extreme paucity
combined treatment. In one such study (Firat et al. of data on the feasibility and effectiveness of radiation
2006), 102 patients with Stage III non-small-cell lung therapy in this setting. Recently, Hayman et al. (2007)
cancer having a Karnofsky Performance Status score embarked on a study with the objective to examine
C70 were retrospectively evaluated for co-morbidity. the utilization patterns of palliative radiation therapy
While all patients received thoracic radiation therapy, among elderly patients with Stage IV non-small-cell
57 (56%) patients also received either sequential or lung cancer and, in particular, to identify factors
concurrent chemotherapy. Multivariate analysis iden- associated with its use. A retrospective population-
tified the use of radiation therapy alone (P \ 0.01) and based cohort study was performed using linked Sur-
presence of grade 4 co-morbidity (P = 0.02) were both veillance, Epidemiology, and End Results-Medicare
independent prognosticators of inferior overall sur- data to identify 11,084 Medicare beneficiaries aged
vival. Contrary to that, Stage IIIB, age C70 years, C65 years who presented with Stage IV non-small-
radiation therapy dose[63 Gy and weight loss of[5% cell lung cancer in the 11 Surveillance, Epidemiology,
were not. Similarly, more recent and retrospective and End Results regions between 1991 and 1996. The
study of Semarau et al. (2008) in 66 inoperable primary outcome was receipt of radiation therapy and
non-small-cell lung cancer patients also confirmed logistic regression analysis was used to identify
independent and adverse influence of cardiac and factors associated with receipt of radiation therapy.
pulmonary co-morbidity on treatment outcome in a A total of 58% of these patients received radiation
multivariate analysis. Finally, Coate et al. (2011) therapy, with its use decreasing over time (P = 0.01).
performed a retrospective review of 740 Stage III Increasing age was negatively associated with receipt
non-small-cell lung cancer patients who were classified of treatment (P \ 0.001), as was increasing co-mor-
by treatment plan: palliative (palliative chemotherapy bidities (p \ 0.001). Factors positively associated
or radiation therapy [B40 Gy]); non-surgical multim- with the receipt of radiation therapy included income
odality ([40 Gy radiation therapy ± chemotherapy); (P = 0.001), hospitalization (P \ 0.001), and treat-
or surgical multimodality (chemotherapy, radiation ment with chemotherapy (P \ 0.001). Although the
therapy, and surgery). Patients [65 years of age were use varied across the Surveillance, Epidemiology, and
more likely to have poor performance status End Results regions (P = 0.001), gender, race/
(P \ 0.0001), multiple co-morbidities (P \ 0.0001), ethnicity, and distance to the nearest radiation therapy
and to receive palliative therapy only (P \ 0.0001). facility were not associated with treatment. The
Older and younger patients treated with curative intent authors concluded that elderly patients with
Radiation Therapy for Lung Cancer in Elderly 533

metastatic non-small-cell lung cancer frequently the initial response rates are high, only approximately
receive palliative radiation therapy, but its use varies, 5% of patients survive 3 years. Of all small-cell lung
especially with age and receipt of chemotherapy. cancer cases, only about 20–30% of all patients pre-
Additional research is needed to determine whether sent with a tumor confined to the hemithorax of ori-
this variability reflects good quality care. gin, the mediastinum or the supraclavicular lymph
In order to define ‘‘optimal’’ treatment approach in nodes, designated as having ‘limited disease’ small-
these patients, Jeremic et al. (1999c) designed and cell lung cancer. All other patients present with dis-
performed a phase II study evaluating short-term seminated, extended disease. Since they have a dismal
chemotherapy given concurrently with palliative prognosis and are, therefore, treated mostly with
radiation therapy. A total of 50 patients entered into a palliative intention, this section focuses on limited
phase II study that used two cycles of carboplatin, disease small-cell lung cancer in elderly.
300 mg/m2, days 1 and 29 and oral etoposide, 50 mg/m2, Importance of active treatment in elderly with
days 1–21 and 29–42. Radiation therapy was admi- limited disease small-cell lung cancer was shown by
nistered with the total radiation dose of 14 Gy given Shepherd et al. (1994) who undertook a retrospective
in 2 fractions administered with 1 week split (days review of elderly patients with small-cell lung cancer
1 and 8). There were 47 patients evaluable for the in an attempt to assess the effect of age on treatment
response, of which there were three (6%) complete decisions, response, survival, and toxicity. There were
responses, and 10 (21%) partial responses. Response 123 patients with age [70 years. There were 74
duration ranged 2–8 months (median, 5 months; patients aged 70–74, 35 aged 75–80, and 14 aged
mean, 5 months). Median survival time-based on 80 years or older. No significant differences existed
intent-to-treat analysis, for all 50 patients was between the groups in sex, stage, performance status,
7 months and 1–3 year survival rates were 31, 4.1, or presence of co-morbid disease. Median survivals
and 2%, respectively. There were nine (19%) patients for patients with limited and extensive disease were
experiencing hematological grade 3 toxicity, and all 11.9 and 5.2 months, respectively (P \ 0.0001), with
other chemotherapy-induced toxicity was either grade no significant difference for patients in any age group
1 or grade 2. Of radiation therapy-induced high-grade (P = 0.4). For both limited and extensive disease,
toxicity (according to the Radiation Therapy Oncol- survival correlated strongly with the treatment
ogy Group), grade 3 esophageal was observed in nine received. Twenty-five patients received no treatment
(19%) patients while only four (9%) patients experi- (median survival time, 1.1 months), 20 had radiation
enced grade 3 bronchopulmonary toxicity. No grade 4 therapy only (median survival time, 7.8 months), and
or 5 toxicity occurred during this study. This study 27 patients had \3 cycles of chemotherapy (median
showed that short-course chemotherapy and palliative survival time, 3.9 months). Median survival time for
radiation therapy in elderly patients with Stage IV the 50 patients who had 4–6 cycles was 10.7 months
non-small-cell lung cancer can be well-tolerated with (limited disease 15.0 months, extensive disease
mild to moderate toxicity. Together with results 8.61 months). Results of this retrospective study
obtained this way, they warrant further studies eval- showed that active treatment should not be withheld
uating the effectiveness of this approach and possible from elderly patients with small-cell lung cancer on
chemotherapy- and/or radiation therapy dose escala- the basis of age. The survival of patients who receive
tion in elderly patients with Stage IV (metastatic) active treatment was significantly longer than that of
non-small-cell lung cancer. untreated patients even though frequent dose reduc-
tions for toxicity may be required. The survival ben-
efit was due to treatment effect and was not due to a
3 Small-Cell Lung Cancer selection bias in the cohort of patients chosen for
therapy. Similarly to this, Noguchi et al. (2011) ret-
Small-cell lung cancer represents 15–20% all lung rospectively studied outcome of 45 patients 80 years
cancers cases. Its chemosensitivity and great meta- or older with small-cell lung cancer, comparing it
static potential makes it a good candidate for che- with those of 38 patients aged 70–79 years. Twenty-
motherapy. However, with chemotherapy alone the four (53%) of the 45 patients were treated with
outcome of small-cell lung cancer is poor. Although combination chemotherapy and/or thoracic radiation
534 B. Jeremić and Ž. Dobrić

therapy, which resulted in significant survival benefit extent these results were biased by eligibility criteria
compared with those left untreated (P \ 0.01). The of the trials which restricted the entry of elderly
main reasons for not administrating anti-cancer patients, since one of meta-analyses showed that the
treatments were advanced age ([85 years), poor survival benefit from thoracic radiation therapy was
performance status, and severe co-morbidities. Med- restricted to younger patients (Warde and Payne
ian survival time and 1-year survival of the treated 1992), possibly due to toxicity. Therefore, careful
patients were 13.0 months and 57% for limited dis- selection of elderly suitable for full dose of radiation
ease and 10.3 months and 40% for extensive disease, therapy and chemotherapy is an important issue.
respectively. Despite a lower chemotherapy dose Randomized phase III studies investigating various
being administered, survivals were similar to those of issues of radiation therapy and chemotherapy in
patients aged 70–79 years. Survival benefit was elderly patients with limited disease small-cell lung
observed even in the treated patients with perfor- cancer are lacking, and the prognostic significance of
mance status 2–3 or a moderate degree of co- age in small-cell lung cancer is not well-defined.
morbidity compared with those left untreated. The While some like Southwest Oncology Group and
frequency of grade 3–4 hematologic toxicities was Cancer and Leukemia Group B had demonstrated an
similar between the two age groups. The authors influence of age in limited disease small-cell lung
concluded that the standard chemotherapy regimen cancer (Spiegelman et al. 1989; Albain et al. 1990),
with or without thoracic radiation therapy seems to be others did not confirm these observations (Osterlind
feasible for patients older than 80 years with small- and Andersen 1986; Sagman et al. 1991). Some had
cell lung cancer, even for those with performance also investigated the relationship between the age and
status 2–3 and/or moderate co-morbidity, although toxicity and outcome among elderly treated with
frequent dose adjustment is necessary. combined thoracic radiation therapy and chemother-
Standard treatment for limited disease small-cell apy in small-cell lung cancer. Findlay et al. (1991)
lung cancer nowadays is combined radiation therapy observed significantly more toxicity in the intensively
and chemotherapy. This practice has been widely treated group (cyclophosphamide, doxorubicin, vin-
accepted after the survival benefit of thoracic radia- cristin) than in the less intensive group of patients
tion therapy has been confirmed by two meta-analyses (single agents, planned dose reductions, or radiation
published in 1992 (Pignon et al. 1992; Warde and therapy alone), which was accompanied by a higher
Payne 1992). The most widely used approach in response rate in the intensively treated group. How-
limited disease small-cell lung cancer consists of four ever, in the subgroup of patients with limited disease
cycles of cisplatin/etoposide and thoracic radiation small-cell lung cancer intensive treatment did not lead
therapy. Due to findings of a meta-analysis demon- to an improved in overall survival.
strating that prophylactic cranial irradiation improves Several studies investigated the influence of age
survival for limited disease small-cell lung cancer among various prognostic factors in small-cell lung
patients in complete remission after radiochemother- cancer (Table 1) (Siu et al. 1996; Dajczman et al.
apy (Auperin et al. 1999), prophylactic cranial irra- 1996; Nou 1996; Jara et al. 1999; Yuen et al. 2000).
diation routinely follows in case of complete As expected, due to various cut-off values used with
remission. There are, however, initiatives to include regard to age as prognostic factor, conflicting results
patients with ‘‘good partial response’’ in the group of were observed. Importantly, in all of the studies fre-
patients suitable for prophylactic cranial irradiation. quent dose emissions/reduced number of chemother-
Importantly, this meta-analysis (Auperin et al. 1999) apy cycles (Siu et al. 1996; Dajczman et al. 1996;
did not show detrimental effect of age on treatment Nou 1996; Yuen et al. 2000) or dose reductions/less
outcome. intensive chemotherapy (Jara et al. 1999) or less
Owing to the lack of specifically designed clinical frequent use of radiation therapy occurred. This
trials addressing this issue in elderly, evidence for the reconfirmed previous observation that elderly was not
standard treatment for the vast majority of elderly only the group receiving less intensive treatment, but
patients having limited disease small-cell lung cancer less likely to be included in clinical trials as well. The
is derived from phase III trials in which elderly are situation seems to remain the same nowadays. In the
largely under represented. It is still unclear to what retrospective analysis of 174 patients with limited
Radiation Therapy for Lung Cancer in Elderly 535

Table 1 Retrospective studies in patients with limited disease small-cell lung cancer treated with radiation therapy and
chemotherapy
Author year Age/n RR (%) survival Toxicity Comments
Siu et al. 1996 \70: 580 78%; n.s. 5 yr.OS: 8%; Only cardiac grade 3/ All LD; CAV/
70: 88 82%; n.s. n.s. 4 toxicity More PE ? TI ? PCI (if CR) age
5 yr.OS: 11%; frequent in patients 70 not a prognostic factor in
n.s. multivariate
Dajczman 1996 \60: LD 49%; n.s. 2 yr. OS: 45 Fewer high-grade N = 123 LD, N = 89 ED;
et al. 45, ED 52%; n.s. 2 yr. OS: 48 toxicity and the mean CAV or PE ? TI;
55 51%; n.s. 2 yr. OS: 43 number of toxicities in No separate analysis for
60–60: elderly LD ? ED [70: only 23%
LD 48, received optimal treatment
ED (compared to 43%/50% in
73 70: the younger groups)
LD 43,
ED 57
Nou 1996 70: 243 All: 72% (n.s.) 5 yr.OS: 5%; No difference between 50% LD 50% ED; (85% of
[70: 110 n.s. 70 and [70 LD and 15% of ED treated
5 yr.OS: 1,3%; with CHT (various) ? TI
n.s.
Jara et al. 1999 \70: 25 46%; n.s. MST: 12.3 mo. No difference Only LD evaluated; PE or
70: 12 50%; n.s. MST: 14.9 mo. between 70 and [70 cPE ? TI
Yuen et al. 2000 \70: 80%; n.s. 5 yr.EFS: 19%; Grade 3/4 All LD, except data for LD
271 88%; n.s. n.s. hematological toxicity (N = 37) ? ED (N = 57)
70: 50 5 yr.EFS: 16%; higher in the elderly
n.s. group, all other
adverse effects: no
difference
Ludbrok 2003 \65: 55 91% MST: 17 mo. No difference in the Age not significant
et al. 65–74: 79% 2 yr OS: 37% incidence of acute or prognosticator in
76 74% MST: 12 mo. late grade 3/4 toxicity multivariate analysis since
[75: 43 2 yr OS: 22% elderly were less frequently
MST: 7 mo. treated with RT/CHT,
2 yr OS: 19% intensive CHT, and PCI
p = 0.003
Schild 2005 \70: 209 n.a 2 yr: 48%;5 yr Of all non-grade 5 Grade 5 toxicity occurred in
et al. [70: 54 n.a : 22% toxicity, only grade 4 6% of those [70 yrs, and in
2 yr: 33%;5 yr pneumonitis more 0.5% of those \ 70 yrs
: 17% frequent in group [70 (p = 0.03)
RR response rates; n.s. not significant; LD limited disease; CAV cyclophosphamide, doxorubicine, vincristin; PE cisplatin, eto-
poside; TI thoracic irradiation; PCI prophylactic cranial irradiation; CR complete response; OS overall survival; ED extensive
disease; MST median survival time; CHT: chemotherapy; cPE carboplatin, etopsoide; Hfx Ac RT hyperfractionated accelerated
radiotherapy

disease small-cell lung cancer, Ludbrok et al. (2003) prognosticator of treatment outcome. It is more than
recently reconfirmed that during the 1990s of the last interesting, however, to observe that none of the
century elderly continued to be under diagnosed and studies observed significantly inferior response rate,
undertreated, resulting in lower median and overall overall or event-free survival for elderly. If therapy is
survival rates. Interestingly, toxicity and pattern of administered, therefore, the outcome in elderly
failure showed no differences when elderly were patients is the same to that observed in non-elderly
compared to their non-elderly counterparts. When, patients. Siu et al. (1996) observed that age influenced
however, multivariate analysis was done in some survival in the univariate analysis, but not in a mul-
studies, age was not shown to be an independent tivariate analysis. It is possible that rather than age
536 B. Jeremić and Ž. Dobrić

itself, factors frequently associated with age, such as patients (Nou 1996; Jara et al. 1999; Yuen et al.
co-morbidity, performance status or less intensive 2000).
treatment may have influenced the prognosis. Few prospective studies (Table 2) specifically
In a re-analysis of original North Central Cancer addressed the issue of optimizing the treatment
Group (Bonner et al. 1999), focused on the influence of approach in elderly patients with limited disease
age on treatment outcome (Schild et al. 2005), a total of small-cell lung cancer. In one such attempt (Westeel
263 patients with limited disease small-cell lung cancer et al. 1998), a regimen of cisplatin, doxorubicin,
and an Eastern Cooperative Oncology Group perfor- vincristine, and etoposide was designed for patients
mance status of B2 who were randomized to receive older than 65 years with small-cell lung cancer and
once-daily radiation therapy or split-course hyper- compared with standard chemotherapy regimens to
fractionated radiation therapy. The outcomes of the 209 maintain efficacy, diminish toxicity, enhance com-
(79%) younger patients (age \70 years old) were pliance, and improve chemotherapy administration
compared with the 54 (21%) elderly patients (age convenience at an acceptable cost. Patients with
C70 years old). Elderly patients were presented with limited-stage disease and selected patients with
significantly greater weight loss and poorer perfor- extensive-stage disease received thoracic radiation
mance status. The 2- and 5-year survival rates were 48 therapy delivered concurrently with cisplatin/etopo-
and 22% for younger patients compared with 33% and side at the time of the second chemotherapy cycle.
17% for older patients (P = 0.14). No difference was There were 25 patients with limited-stage disease and
also found when using local control, distant metastasis 41 patients with extensive-stage disease. Median
control and freedom from progression. Grade C4 survival was 70 weeks and 5-year survival was 25%
pneumonitis occurred in 0% of those patients age for limited-stage disease. Only one treatment-related
\70 years compared with 6% of older patients death occurred and severe toxicity was infrequent.
(P = 0.008). Grade 5 toxicity occurred in 1 of 209 The median delivered dose-intensity was according to
(0.5%) patients age\70 years compared with three of protocol and the mean delivered total dose was 80%
54 (5.6%) older patients (P = 0.03). Despite having of intended. The treatment outcome achieved with
more weight loss, poorer performance status, increased this treatment approach in a phase II study of elderly
pulmonary toxicity, and more deaths due to treatment, patients compared favorably with published results of
survival was not found to be significantly worse in older standard regimens in patient populations with better
individuals. Fit elderly patients with limited disease prognostic factors. Murray et al. (1998) tailored an
small-cell lung cancer can receive combined-modality approach for elderly, infirm or non-compliant patients
therapy with the expectation of relatively favorable using only two cycles of chemotherapy (cyclophos-
long-term survival. phamide/doxorubicin/vincristin and cisplatin/etopo-
This was also an observation of several investi- side) and radiation therapy (20 Gy in 5 fractions or
gators reporting on institutional experience in limited 30 Gy in 10 fractions). Toxicity was low, except three
number of patients (Shimizu et al. 2007; Okamoto treatment-related deaths, two of which were due to
et al. 2010) treated with concurrent radiation therapy cardiac toxicity, with likely ischemic cause. The
and chemotherapy. Median progression-free survivals median time to progression was 40 weeks and 2-year
of approximately 15 months and median overall sur- progression-free survival was 25%. The median sur-
vivals of approximately 25 months have been vival time was 54 weeks and 5-year survival rate was
achieved with acceptable toxicity. 18%. The median survival time and 5-year survival
Some studies also investigated the influence of age were similar for 18 patients \70 years and for the 37
on toxicity. While some observed fewer elderly patients C70 years. In another study, Jeremic et al.
patients with high-grade toxicity (P = 0.0001), and (1998a) also administered only two courses of che-
similar incidence of treatment-related deaths due to motherapy (carboplatin, 400 mg/m2, days 1 and 29
less intensive treatment (Dajczman et al. 1996), other and oral etoposide 50 mg/m2, days 1–21 and 29–49)
reported on similar toxicities in both age groups. In concurrently with accelerated hyperfractionated
studies where a higher rate of hematological toxicity radiation therapy (45 Gy in 30 fractions in 15 treat-
and fatal toxicities occurred in the elderly group, ment days using 1.5 Gy b.i.d. fractionation) in 75
other toxicities were similar compared to non-elderly patients C70 years with a Karnofsky performance
Radiation Therapy for Lung Cancer in Elderly 537

Table 2 Phase II studies of radiation therapy and chemotherapy in elderly and/or frail patients with LD-SCLC
AUTHOR (yr) N CHT RT (Gy/fx) N Age [ 70 (yrs) (%) PS [ 2 (%)
Westeel et al. (1998) 25 PAVEx3, PEx1 20/5 25 med. 72 28
30/10
40/5
Murray et al. (1998) 55 CAVx1, PEx1 20/5 55 67 45
30/10
Jeremic et al. (1998a) 72 CBDCA/oral Ex2 45/30 72 100 17
(BID)
Matsui et al. (1998) 16 CBDCA/oral Ex4 45/24 16 100 n.a.
CHT chemotherapy, RT radiation therapy, Gy Grey, fx fraction, PS performance status; not available, P cisplatin, A doxorubicin,
V vincristine, E etoposide, C cyclophosphamide, CBDCA carboplatin, BID two fractions a day

status score of [60% and with no major concomitant What summary of retrospective and prospective
diseases. The median survival time was 15 months studies also showed is a similar outcome of elderly
and 5-year survival was 13%. Good pre-treatment and non-elderly patients with limited disease small-
patient characteristics led to 83% patients receiving cell lung cancer, despite elderly frequently receiving
therapy on an outpatient basis and low toxicity. Grade less-intensive chemotherapy and/or thoracic radiation
4 thrombocytopenia occurred in 1.4% of all patients, therapy. Additionally, despite less compliance in
thrombocytopenia grade 3 in 11%, grade 3 leucopenia elderly, no difference in either response rates or sur-
in 8.3%, grade 3 anemia in 2.8%, infection in 4.2%, vival was detected between them and their non-
and nausea and vomiting in 4.2% of all patients. No elderly counterparts (Kelly et al. 1991; Siu et al. 1996;
high-grade bronchopulmonary toxicity was observed Dajczman et al. 1996; Tebbutt et al. 1997; Yuen et al.
and grade 3 esophagitis occurred in only 2.8% of the 2000). Although the reason for this finding is still
patients. Additional advantage of this approach was unclear, possible explanation may lie in a different
its short duration, resulting in more time spent at metabolism of drugs. This may lead to a need for
home, and therefore, a good quality of life. In lower doses of various drugs in elderly (Montamat
addition to these two studies, Matsui et al. (1998) et al. 1989; McKenna 1994; Joss et al. 1995), since
reported on 16 patients with limited disease small- different biological behavior of tumors in elderly is
cell lung cancer [70 years for whom four cycles of not very likely cause of this observation (Matsui et al.
carboplatin and oral etoposide (40 mg/m2, days 1998). As pointed out (Yuen et al. 2000), there may
1–14) were followed by chest radiation therapy be a threshold above which a significant benefit can
(45 Gy). The median survival time was 15.1 months be realized. The delivery of ‘‘adequate enough’’
and a 2 year survival rate was 21.8%. For patients treatment to achieve a positive effect could, perhaps,
C75 years the median survival time was still be achieved with the modest dose reductions.
10.3 months and 2 year survival rate was 11.3%. This threshold will be hard to specify and document,
Grade 3 and 4 leucopenia occurred in 36 and 14% of but it seems that studies of Jeremic et al. (1998b) and
patients, respectively, and grade 3 and 4 thrombo- Murray et al. (1998) supported this statement:
cytopenia occurred in 39 and 14% of the patients, although chemotherapy was limited to only two
respectively. Grade 3/4 anemia occurred in 50% of cycles given concurrently with thoracic radiation
patients. Non-hematological toxicity was rare. What therapy, it was possible to obtain results which are not
these three prospective studies have shown is that substantially inferior to those obtained with more
well-tailored treatment approaches, in a well-selected intensive approaches. However, every caution should
patient population, carefully balancing between be suggested in this patient population, particularly
‘‘optimal’’ thoracic radiation therapy and chemother- regarding hematological toxicity.
apy elderly can tolerate while avoiding unnecessary In extensive disease small-cell lung cancer, stan-
toxicity, may lead to high treatment success and dard treatment for patients with extensive disease
toxicity profile not very different to that usually small-cell lung cancer is chemotherapy. The addition
observed in non-elderly patients. of thoracic radiation therapy did not improve survival
538 B. Jeremić and Ž. Dobrić

in the past, and thoracic radiation therapy was applied accumulated evidence clearly show that ‘‘fit’’ elderly
only for palliation of local symptoms when chemo- may tolerate the treatments considerably well, regard-
therapy alone was not efficient (Livingston et al. less of its intensity. The starting point may well be
1984). However, a recent prospective randomized inclusion of more of elderly in clinical studies as well as
trial by Jeremic et al. (1999d) showed an advantage designing of specific clinical trials exclusively dedi-
for three cycles of platinum-etoposide chemotherapy cated to elderly. In addition, every caution should be
followed by accelerated hyperfractionated thoracic undertaken in order not to over emphasize the results of
radiation therapy given concurrently with low-dose, recent studies which are, unfortunately, mostly retro-
daily carboplatin/etoposide over chemotherapy with spective. It said that, inherent biases and underlying
platinum/etoposide (five cycles) alone. Important problems, unsolved so far, may hamper future
finding of that study was survival advantage being endeavors having the same goal: enabling elderly with
observed for patients [60 years old. This finding was lung cancer equal diagnostic and treatment approach as
confirmed by the multivariate analysis identifying the their younger counterparts receive, on or off the pro-
age as an independent prognosticator of survival in tocol. While this should be continuous reminder to all
patients with extensive disease small-cell lung cancer wishing to engage in this setting, definite solution
(unpublished observations; drawn from Jeremic et al. remains within the domain of more clinical studies in
1999d). elderly with lung cancer and we need it now.

4 Conclusions References
Accumulated evidence identifies radiation therapy as
Ahmad E, Sandhu AP, Fuster MM, Messer K, Pu M, Nobiensky P,
an important treatment modality in elderly patients Bazhenova L, Seagren S (2010) Hypofractionated radiother-
with lung cancer. This is so irrespective of the con- apy as definitive treatment of Stage I non-small-cell lung
sideration of cut-off age, histology or stage or whether cancer in older patients. Am J Clin Oncol. 17 Jun 2010. [Epub
radiation therapy had been used alone or in combi- ahead of print]
Albain KS, Crowley JJ, LeBlanc M, Livingston RB (1990)
nation with chemotherapy. Encouraging results were Determinants of improved outcome in small-cell lung
obtained in both non-small-cell lung cancer and cancer: an analysis of the 2,580-patient Southwest Oncology
small-cell lung cancer, although prospective studies, Group data base. J Clin Oncol 8:1563–1574
especially randomized ones are basically lacking. Aristizabal SA, Caldwell WL (1976) Radical irradiation with
the split-course technique in carcinoma of the lung. Cancer
Unfortunately, current evidence also points out 37:2630–2635
that age alone is an uncertain prognostic criterion Aristizabal SA, Meyerson M, Caldwell WL, Mayer EG (1976)
when outcome including toxicity is considered. Bio- Age as a prognostic indicator in carcinoma of the lung.
logical age of each individual elderly seems to be Radiology 121:721–723
Atagi S, Kawahara M, Ogawara M, Matsui K, Masuda N,
more important than chronological age. This requires Kudoh S, Negoro S, Furuse K (2000) Phase II trial of daily
a specific geriatric assessment of each individual low-dose carboplatin and thoracic radiotherapy in elderly
patient, including detection of co-morbidities and the patients with locally advanced non-small-cell lung cancer.
functional capacity for performing activities of daily Jpn J Clin Oncol 30:59–64
Atagi S, Kawahara M, Tamura T, Noda K, Watanabe K,
living, the cognitive and nutritional status of the Yokoyama A, Sugiura T, Senba H, Ishikura S, Ikeda H,
patient. The decision- making of optimal treatment in Ishizuka N, Saijo N, Japan Clinical Oncology Group
elderly patients with lung cancer should be based on (JCOG9812) (2005) Standard thoracic radiotherapy with
both disease and patient-specific criteria. Age itself is or without concurrent daily low-dose carboplatin in elderly
patients with locally advanced non-small-cell lung cancer: a
not a contraindication for applying the standard phase III trial of the Japan Clinical Oncology Group
treatment. However, the individualized management (JCOG9812). Jpn J Clin Oncol 35:195–201
of the elderly must reflect the results of a compre- Auperin A, Arriagada R, Pignon JP, Gregor A, Stephens RJ,
hensive geriatric assessment. Kristjansen PE, Johnson BE, Ueoka H, Wagner H, Aisner J
(1999) Prophylactic cranial irradiation for patients with
Major issue in this field remains the lack of pro- small-cell lung cancer in complete remission. Prophylactic
spective clinical studies investigating ‘‘optimal’’ cranial irradiation overview collaborative group. N Engl J
treatments in this setting. This is especially so, since Med 341:476–484
Radiation Therapy for Lung Cancer in Elderly 539

Bach PB, Cramer LD, Warren JL, Begg CB (1999) Racial fractionated accelerated radiotherapy (CHART) in patients
differences in the treatment of early-stage lung cancer. aged [70 years with Stage 1 non-small-cell lung cancer.
N Engl J Med 341:1198–1205 Eur J Cardiothorac Surg 24:1002–1007
Balducci L, Extermann M (2000a) Cancer and aging. An Giorgo CG, Pappalardo A, Ruso A (2007) A phase II induction
evolving panorama. Hematol Oncol Clin North Am 14:1–16 chemotherapy followed by concurent chemoradiotherapy in
Balducci L, Extermann M (2000b) Management of cancer in the elderly patients with locally advanced non-small-cell lung
older person: a practical approach. Oncologist 5:224–237 cancer. Anticancer Drugs 18:713–719
Bernabei R, Venturiero R, Tarsitani P, Gambassi G (2000) The Haasbeek CJ, Lagerwaard FJ, Antonisse ME, Slotman BJ,
comprehensive geriatric assessment: When, where, how. Senan S (2010) Stage I non-small-cell lung cancer in
Crit Rev Oncol Hematol 33:45–56 patients aged [ or =75 years: Outcomes after stereotactic
Bonner JA, Sloan JA, Shanahan TG, Brooks BJ, Marks RS, radiotherapy. Cancer 116:406–414
Krook JE, Gerstner JB, Maksymiuk A, Levitt R, Mailliard JA, Hanna N, Neubauer M, Yiannoutsos C, McGarry R, Arseneau J,
Tazelaar HD, Hillman S, Jett JR (1999) Phase III comparison Ansari R, Reynolds C, Govindan R, Melnyk A, Fisher W,
of twice-daily split-course irradiation versus once-daily Richards D, Bruetman D, Anderson T, Chowhan N, Nattam S,
irradiation for patients with limited stage small-cell lung Mantravadi P, Johnson C, Breen T, White A, Einhorn L,
carcinoma. J Clin Oncol 17:2681–2691 Hoosier Oncology Group; US Oncology (2008) Phase III
Clamon G, Herndon J, Cooper R, Chang AY, Rosenman J, study of cisplatin, etoposide, and concurrent chest radiation
Green MR (1999) Radiosensitization with carboplatin for with or without consolidation docetaxel in patients with
patients with unresectable Stage III non-small-cell lung inoperable Stage III non-small-cell lung cancer: the Hoosier
cancer: a phase III trial of the Cancer and Leukemia Group Oncology Group and U.S. Oncology. J Clin Oncol 26:
B and the Eastern Cooperative Oncology Group. J Clin 5755–5760
Oncol 17:4–11 Hayakawa K, Mitsuhashi N, Saito Y, Nakayama Y, Furuta M,
Coate LE, Massey C, Hope A, Sacher A, Barrett K, Pierre A, Sakurai H, Kawashima M, Ohno T, Nasu S, Niibe H (1999)
Leighl N, Brade A, de Perrot M, Waddell T, Liu G, Feld R, Limited field irradiation for medically inoperable patients
Burkes R, Cho BC, Darling G, Sun A, Keshavjee S, Bezjak A, with peripheral Stage I non-small-cell lung cancer. Lung
Shepherd FA (2011) Treatment of the elderly when cure is the Cancer 26:137–142
goal: the influence of age on treatment selection and efficacy Hayakawa K, Mitsuhashi N, Katano S, Saito Y, Nakayama Y,
for Stage III non-small-cell lung cancer. J Thorac Oncol Sakurai H, Akimoto T, Hasegawa M, Yamakawa M, Niibe H
6:537–544 (2001) High-dose radiation therapy for elderly patients with
Coy P, Kennelly GM (1980) The role of curative radiotherapy inoperable or unresectable non-small-cell lung cancer. Lung
in the treatment of lung cancer. Cancer 45:698–702 Cancer 32:81–88
Dajczman E, Fu LY, Small D, Wolkove N, Kreisman H (1996) Hayman JA, Abrahamse PH, Lakhani I, Earle CC, Katz SJ
Treatment of small-cell lung carcinoma in the elderly. (2007) Use of palliative radiotherapy among patients with
Cancer 77:2032–2038 metastatic non-small-cell lung cancer. Int J Radiat Oncol
Extermann M (2000) Measuring co-morbidity in older cancer Biol Phys 69:1001–1007
patients. Eur J Cancer 36:453–471 Higton AM, Monach J, Congleton J (2010) Investigation and
Findlay MP, Griffin AM, Raghavan D, McDonald KE, Coates management of lung cancer in older adults. Lung Cancer
AS, Duval PJ, Gianoutsos P (1991) Retrospective review of 69:209–212
chemotherapy for small-cell lung cancer in the elderly: Jara C, Gomez-Aldaravi JL, Tirado R, Meseguer VA, Alonso C,
Does the end justify the means? Eur J Cancer 27:1597–1601 Fernandez A (1999) Small-cell lung cancer in the elderly: is
Firat S, Pleister A, Wirad RW, Gore E (2006) Age is independent age of patient a relevant factor? Acta-Oncol 38:781–786
of comorbidity influencing patient selection for combined Jatoi A, Schild SE, Foster N, Henning GT, Dornfeld KJ, Flynn
modality therapy for treatment of Stage III non-small-cell PJ, Fitch TR, Dakhil SR, Rowland KM, Stella PJ, Soori GS,
lung cancer (NSCLC). Am J Clin Oncol 29:252–257 Adjei AA (2010) A phase II study of cetuximab and
Furuse K, Fukuoka M, Kawahara M, Nishikawa H, Takada Y, radiation in elderly and/or poor performance status patients
Kudoh S, Katagami N, Ariyoshi Y (1999) Phase III study of with locally advanced non-small-cell lung cancer (N0422).
concurrent versus sequential thoracic radiotherapy in com- Ann Oncol 21:2040–2044
bination with mitomycin, vindesine, and cisplatin in unre- Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1997)
sectable Stage III non-small-cell lung cancer. J Clin Oncol Hyperfractionated radiotherapy alone for clinical Stage I
17:2692–2699 non-small-cell lung cancer. Int J Radiat Oncol Biol Phys
Gauden SJ, Tripcony L (2001) The curative treatment by 38:521–525
radiation therapy alone of Stage I non-small-cell lung Jeremic B, Shibamoto Y, Milicic B, Nikolic N, Dagovic A,
cancer in a geriatric population. Lung Cancer 32:71–79 Milisavljevic S (1998a) Concurrent radiochemotherapy for
Gauden S, Ramsay J, Tripcony L (1995) The curative treatment patients with Stage III non-small-cell lung cancer (NSCLC):
by radiotherapy alone of Stage I non-small cell carcinoma long-term results of a phase II study. Int J Radiat Oncol Biol
of the lung. Chest 108:1278–1282 Phys 42:1091–1096
Gava A (1999) Lung cancer radiation treatment in the elderly. Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1998b)
Crit Rev Oncol Hematol 32:45–48 Carboplatin, etoposide, and accelerated hyperfractionated
Ghosh S, Sujendran V, Alexiou C, Beggs L, Beggs D (2003) radiotherapy for elderly patients with limited small-cell lung
Long term results of surgery versus continuous hyper carcinoma: a phase II study. Cancer 82:836–841
540 B. Jeremić and Ž. Dobrić

Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1999a) Ludbrok JJ, Truong PT, MacNeil MV, Lesperance M, Webber A,
Hyperfractionated radiotherapy for clinical Stage II non- Joe H, Martins H, Lim J (2003) Do age and co-morbidity
small-cell lung cancer. Radiother Oncol 51:141–145 impact treatment allocation and outcomes in limited Stage
Jeremic B, Shibamoto Y, Milicic B, Milisavljevic S, Nikolic N, small-cell lung cancer? A community-based population
Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999b) analysis. Int J Radiat Oncol Biol Phys 55:1321–1330
A phase II study of concurrent accelerated hyperfraction- Matsui K, Masuda N, Fukuoka M, Yana T, Hirashima T,
ated radiotherapy and carboplatin/oral etoposide for elderly Komiya T, Kobayashi M, Kawahara M, Atagi S, Ogawara
patients with Stage III non-small-cell lung cancer. Int J M, Negoro S, Kudoh S, Furuse K (1998) Phase II trial of
Radiat Oncol Biol Phys 44:343–348 carboplatin plus oral etoposide for elderly patients with
Jeremic B, Shibamoto Y, Milicic B, Milisavljevic S, Nikolic N, small-cell lung cancer. Br J Cancer 77:1961–1965
Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999c) McKenna RJ Sr (1994) Clinical aspects of cancer in the elderly.
Short-term chemotherapy and palliative radiotherapy for Treatment decisions, treatment choices, and follow-up.
elderly patients with Stage IV non-small-cell lung cancer. A Cancer 74:2107–2117
phase II study. Lung Cancer 24:1–9 Monfardini S, Sorio R, Boes G, Kaye S, Serraino D (1995)
Jeremic B, Shibamoto Y, Nikolic N, Milicic B, Milisavljevic S, Entry and evaluation of elderly patients in European
Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999d) Organization for research and Treatment of Cancer (EO-
Role of radiation therapy in the combined-modality treat- RTC) new-drug-development studies. Cancer 76:333–338
ment of patients with extensive disease small-cell lung Montamat SC, Cusack BJ, Vestal RE (1989) Management of
cancer: a randomized study. J Clin Oncol 17:2092–2099 drug therapy in the elderly. N Engl J Med 321:303–309
Joss RA, Bacchi M, Hurny C, Bernhard J, Cerny T, Martinelli Montella M, Gridelli C, Crispo A, Scognamiglio F, Ruffolo P,
G, Leyvraz S, Senn HJ, Stahel R, Siegenthaler P (1995) Gatani T, Boccia V, Maione P, Fabbrocini G (2002) Has
Early versus late alternating chemotherapy in small-cell lung cancer in the elderly different characteristics at
lung cancer. Swiss Group for Clinical Cancer Research presentation? Oncol Rep 9:1093–1096
(SAKK). Ann Oncol 6:157–166 Morita K, Fuwa N, Suzuki Y, Nishio M, Sakai K, Tamaki Y,
Karasawa K, Niibe Y, Igaki H, Ieki R, Tanaka Y (2002) Niibe H, Chujo M, Wada S, Sugawara T, Kita M (1997)
Radiotherapy combined with bronchial arterial infusion of Radical radiotherapy for medically inoperable non-small-
CDDP in the treatment of Stage III non-small-cell lung cell lung cancer in clinical Stage I: a retrospective analysis
cancer for aged patients. Int J Radiation Oncol Biol 54 of 149 patients. Radiother Oncol 42:31–36
(Suppl): 104 (Abstract 175) Movsas B, Scott C, Sause W, Byhardt R, Komaki R, Cox J,
Kaskowitz L, Graham MV, Emami B, Halverson KJ, Rush C Johnson D, Lawton C, Dar AR, Wasserman T, Roach M,
(1993) Radiation therapy alone for Stage I non-small-cell Lee JS, Andras E (1999) The benefit of treatment intensi-
lung cancer. Int J Radiat Oncol Biol Phys 27:517–523 fication is age and histology-dependent in patients with
Kelly P, O’Brien AA, Daly P, Clancy L (1991) Small-cell lung locally advanced non-small-cell lung cancer (NSCLC): A
cancer in elderly patients: the case for chemotherapy. Age quality-adjusted survival analysis of radiation therapy
Ageing 20:19–22 oncology group (RTOG) chemoradiation studies. Int J
Krol AD, Aussems P, Noordijk EM, Hermans J, Leer JW Radiat Oncol Biol Phys 45:1143–1149
(1996) Local irradiation alone for peripheral Stage I lung Murray N, Grafton C, Shah A, Gelmon K, Kostashuk E, Brown
cancer: Could we omit the elective regional nodal irradi- E, Coppin C, Coldman A, Page R (1998) Abbreviated
ation? Int J Radiat Oncol Biol Phys 34:297–302 treatment for elderly, infirm, or noncompliant patients with
Kusumoto S, Koga K, Tsukino H, Nagamachi S, Nishikawa K, limited-stage small-cell lung cancer. J Clin Oncol 16:
Watanabe K (1986) Comparison of survival of patients with 3323–3328
lung cancer between elderly (greater than or equal to 70) Nakano K, Hiramoto T, Kanehara M, Doi M, Furonaka O,
and younger (70 greater than) age groups. Jpn J Clin Oncol Miyazu Y, Hada Y (1999) Radiotherapy alone for elderly
16:319–323 patients with Stage III non-small-cell lung cancer. Nihon
Langer CJ, Scott CB, Byhardt RW (2000) Effect of advanced Kokyki Gakkai Zasshi 37:276–281
age on outcome in radiation therapy oncology group studies Nakano K, Yamamoto M, Iwamoto H, Hiramoto T (2003)
of locally advanced NSCLC. Lung Cancer 29:104 (Abstract Daily low-dose cisplatin plus concurrent high-dose thoracic
340) radiotherapy in elderly patients with locally advanced
Langer CJ, Hsu C, Curran WJ Jr et al (2001) Do elderly unresectable non-small-cell lung cancer. Gan To Kagaku
patients with locally advanced non-small-cell lung cancer Ryoho 30:1283–1287
benefit from combined modality therapy? A secondary Newaishy GA, Kerr GR (1989) Radical radiotherapy for
analysis of RTOG 94–10. Int J Radiat Oncol Biol Phys bronchogenic carcinoma: five year survival rates. Clin
51(Suppl 23):31 Oncol (R Coll Radiol) 1:80–85
Livingston RB, Mira JG, Chen TT, McGowan M, Costanzi JJ, Niibe Y, Karasawa K, Shibuya M, Ieki R, Tanaka Y (2003)
Samson M (1984) Combined modality treatment of exten- Prospective study of three-dimensional radiation therapy (3D-
sive small-cell lung cancer: a Southwest Oncology Group CRT) using a middle fraction size for small-sized lung tumor in
study. J Clin Oncol 2:585–590 elderly patients. Proc Am Soc Clin Oncol (Abstract 2702)
Lonardi F, Coeli M, Pavanato G, Adami F, Gioga G, Noguchi T, Mochizuki H, Yamazaki M, Kawate E, Suzuki Y,
Campostrini F (2000) Radiotherapy for non-small-cell lung Sato T, Takahashi H (2010) A retrospective analysis of
cancer in patients aged 75 and over: Safety, effectiveness clinical outcomes of patients older than or equal to 80 years
and possible impact on survival. Lung Cancer 28:43–50 with small cell lung cancer. J Thorac Oncol 5:1081–1087
Radiation Therapy for Lung Cancer in Elderly 541

Noordijk EM, vd Poest Clement E, Hermans J, Wever AM, Rosenthal SA, Curran WJ Jr, Herbert SH, Hughes EN,
Leer JW (1988) Radiotherapy as an alternative to surgery in Sandler HM, Stafford PM, McKenna WG (1992) Clinical
elderly patients with resectable lung cancer. Radiother Stage II non-small cell lung cancer treated with radiation
Oncol 13:83–89 therapy alone. The significance of clinically staged ipsilat-
Nou E (1996) Full chemotherapy in elderly patients with small eral hilar adenopathy (N1 disease). Cancer 70:2410–2417
cell bronchial carcinoma. Acta-Oncol 35:399–406 Rubenstein LZ (1995) An overview of comprehensive geriatric
Nugent WC, Edney MT, Hammerness PG, Dain BJ, Maurer LH, assessment: Rationale, history, program models, basic
Rigas JR (1997) Non-small cell lung cancer at the extremes of components. In: Rubinstein LZ, Wieland D, Bernabei R
age: Impact on diagnosis and treatment. Ann Thorac Surg (eds) Geriatric assessment technology: the state of the art.
63:193–197 Springer, New York
Okamoto K, Okamoto I, Takezawa K, Tachibana I, Fukuoka M, Sagman U, Feld R, Evans WK, Warr D, Shepherd FA, Payne D,
Nishimura Y, Nakagawa K (2010) Cisplatin and etoposide Pringle J, Yeoh J, DeBoer G, Malkin A (1991) The
chemotherapy combined with early concurrent twice-daily prognostic significance of pre-treatment serum lactate
thoracic radiotherapy for limited-disease small-cell lung dehydrogenase in patients with small-cell lung cancer.
cancer in elderly patients. Jpn J Clin Oncol 40:54–59 J Clin Oncol 9:954–961
Osterlind K, Andersen PK (1986) Prognostic factors in small San José S, Arnaiz MD, Lucas A, Navarro V, Serrano G,
cell lung cancer: Multivariate model based on 778 patients Zaderazjko M, Jeremic B, Guedea F (2005) Radiation
treated with chemotherapy with or without irradiation. therapy alone in elderly with early stage non-small-cell lung
Cancer Res 46:4189–4194 cancer. Lung Cancer 52:149–154
Osterweil D, Brummel-Smith K, Beck JC (eds) (2000) Com- Sandler HM, Curran WJ Jr, Turrisi AT 3rd (1990) The
prehensive geriatric assessment. McGraw Hill, New York influence of tumor size and pre-treatment staging on
Owonikoko TK, Ragin CC, Belani CP, Oton AB, Gooding WE, outcome following radiation therapy alone for Stage I
Taioli E, Ramalingam SS (2007) Lung cancer in elderly non-small-cell lung cancer. Int J Radiat Oncol Biol Phys
patients: an analysis of the surveillance, epidemiology, and 19:9–13
end results database. J Clin Oncol 25:5570–5577 Schaake-Koning C, van den Bogaert W, Dalesio O, Festen J,
Palma DA, Tyldesley S, Sheehan F, Mohamed IG, Smith S, Hoogenhout J, van Houtte P, Kirkpatrick A, Koolen M,
Wai E, Murray N, Senan S (2010a) Stage I non-small cell Maat B, Nijs A (1992) Effects of concomitant cisplatin and
lung cancer (NSCLC) in patients aged 75 years and older: radiotherapy on inoperable non-small-cell lung cancer.
Does age determine survival after radical treatment? J N Engl J Med 326:524–530
Thorac Oncol. 5:818–824 Schild SE, Stella PJ, Geyer SM, Bonner JA, McGinnis WL,
Palma D, Visser O, Lagerwaard FJ, Belderbos J, Slotman BJ, Mailliard JA, Brindle J, Jatoi A, Jett JR (2003) The outcome
Senan S (2010b) Impact of introducing stereotactic lung of combined-modality therapy for Stage III non-small-cell
radiotherapy for elderly patients with Stage I non-small-cell lung cancer in elderly. J Clin Oncol 21:3201–3206
lung cancer: a population-based time-trend analysis. J Clin Schild SE, Stella PJ, Brooks BJ, Mandrekar S, Bonner JA,
Oncol 28:5153–5159 McGinnis WL, Mailliard JA, Krook JE, Deming RL, Adjei
Park CH, Bonomi M, Cesaretti J, Neugut AI, Wisnivesky JP AA, Jatoi A, Jett JR (2005) Results of combined-modality
(2010) Effect of radiotherapy planning complexity on therapy for limited-stage small-cell lung carcinoma in the
survival of elderly patients with unresected localized lung elderly. Cancer 103:2349–2354
cancer. Int J Radiat Oncol Biol Phys. 2010 Oct 5. [Epub Schild SE, Mandrekar SJ, Jatoi A, McGinnis WL, Stella PJ,
ahead of print] Deming RL, Jett JR, Garces YI, Allen KL, Adjei AA, For
Patterson CJ, Hocking M, Bond M, Teale C (1998) Retrospec- the North Central Cancer Treatment Group (2007) The
tive study of radiotherapy for lung cancer in patients aged value of combined-modality therapy in elderly patients with
75 years and over. Age Ageing 27:515–518 Stage III non-small-cell lung cancer. Cancer 110:363–368
Pergolizzi S, Santacaterina A, De Renzis C, Settineri N, Gaeta M, Semarau S, Klautke G, Wirchow JC, Kundt G, Fietkau R
Frosina P, Russi EG, Altavilla G (2002) Older people with (2008) Impact of comorbidity and age on the outcome of
non small cell lung cancer in clinical Stage IIIA and co- patients with inoperable NSCLC treated with concurrent
morbid conditions. Is curative irradiation feasible? Final chemoradiotherapy. Respir Med 102:210–218
results of a prospective study. Lung Cancer 37:201–206 Sgroi MM, Neubauer M, Ansari R (2007) An analysis of
Pignon JP, Arriagada R, Ihde DC, Johnson DH, Perry MC, elderly patients (pts) treated on a phase III trial of cisplatin
Souhami RL, Brodin O, Joss RA, Kies MS, Lebeau B plus etoposide (E) with concurrent radiotherapy (CRT)
(1992) A meta-analysis of thoracic radiotherapy for small- followed by docetaxel (D) vs observation in patients with
cell lung cancer. N Engl J Med 327:1618–1624 Stage III non-small-cell lung cancer (NSCLC). Proc Am
Pignon T, Gregor A, Schaake-Koning C, Roussel A, Van Soc Clin Oncol 25: (Abstract 9037)
Glabbeke M, Scalliet P (1998) Age has no impact on acute Shepherd FA, Amdemichael E, Evans WK, Chalvardjian P,
and late toxicity of curative thoracic radiotherapy. Radio- Hogg-Johnson S, Coates R, Paul K (1994) Treatment of
ther Oncol 46:239–248 small-cell lung cancer in the elderly. J Am Geriatr Soc
Rocha Lima CM, Herndon JE, Kosty M, Clamon G, Green MR 42:64–70
(2002) Therapy choices among older patients with ling Shimizu T, Sekine I, Sumi M, Ito Y, Yamada K, Nokihara H,
carcinoma: an evaluation of two trials of the Cancer and Yamamoto N, Kunitoh H, Ohe Y, Tamura T (2007)
Leukaemia Group B. Cancer 94:181–187 Concurrent chemoradiotherapy for limited-disease small-cell
542 B. Jeremić and Ž. Dobrić

lung cancer in elderly patients aged 75 years or older. Jpn J Westeel V, Murray N, Gelmon K, Shah A, Sheehan F,
Clin Oncol 37:181–185 McKenzie M, Wong F, Morris J, Grafton C, Tsang V,
Sibley GS, Jamieson TA, Marks LB, Anscher MS, Prosnitz LR Goddard K, Murphy K, Parsons C, Amy R, Page R (1998)
(1998) Radiotherapy alone for medically inoperable Stage I New combination of the old drugs for elderly patients with
non-small-cell lung cancer: the Duke experience. Int J small-cell lung cancer: a phase II study of the PAVE
Radiat Oncol Biol Phys 40:149–154 regimen. J Clin Oncol 16:1940–1947
Siu LL, Shepherd FA, Murray N, Feld R, Pater J, Zee B (1996) Wisnivesky JP, Bonomi M, Henschke C, Iannuzzi M, McGinn
Influence of age on the treatment of limited-stage small-cell T (2005) Radiation therapy for the treatment of unresected
lung cancer. J Clin Oncol 14:821–828 Stage I–II non-small-cell lung cancer. Chest 128:1461–1467
Slotman BJ, Njo KH, Karim AB (1994) Curative radiotherapy Wisnivesky JP, Halm E, Bonomi M, Powell C, Bagiella E
for technically operable Stage I non-small-cell lung cancer. (2010) Effectiveness of radiation therapy for elderly patients
Int J Radiat Oncol Biol Phys 29:33–37 with unresected Stage I and II non-small-cell lung cancer.
Spiegelman D, Maurer LH, Ware JH, Perry MC, Chahinian AP, Am J Respir Crit Care Med 181:264–269
Comis R, Eaton W, Zimmer B, Green M (1989) Prognostic Wurschmidt F, Bunemann H, Bunemann C, Beck-Bornholdt
factors in small-cell carcinoma of the lung: an analysis of 1, HP, Heilmann HP (1994) Inoperable non-small-cell lung
521 patients. J Clin Oncol 7:344–354 cancer: a retrospective analysis of 427 patients treated with
Tebbutt NC, Snyder RD, Burns WI (1997) An analysis of the high-dose radiotherapy. Int J Radiat Oncol Biol Phys
outcomes of treatment of small-cell lung cancer in the 28:583–588
elderly. Aust N Z J Med 27:160–164 Yanick R (1997) Cancer burden in the aged: an epidemiologic
Tombolini V, Bonanni A, Donato V, Raffetto N, Santarelli M, and demographic overview. Cancer 80:1273–1283
Valeriani M, Enrici RM (2000) Radiotherapy alone in elderly Yu HM, Liu YF, Yu JM, Liu J, Zhao Y, Hou M (2008)
patients with medically inoperable Stage IIIA and IIIB non- Involved-field radiotherapy is effective for patients 70 years
small-cell lung cancer. Anticancer Res 20:4829–4833 old or more with early stage non-small-cell lung cancer.
Warde P, Payne D (1992) Does thoracic irradiation improve Radiother Oncol 87:29–34
survival and local control in limited-stage small-cell Yuen AR, Zou G, Turrisi AT, Sause W, Komaki R, Wagner H,
carcinoma of the lung? A meta-analysis. J Clin Oncol Aisner SC, Livingston RB, Blum R, Johnson DH (2000)
10:890–895 Similar outcome of elderly patients in intergroup trial 0096:
Werner-Wasik M, Scott C, Cox JD, Sause WT, Byhardt RW, Cisplatin, etoposide, and thoracic radiotherapy administered
Asbell S, Russell A, Komaki R, Lee JS (2000) Recursive once or twice daily in limited stage small-cell lung
partitioning analysis of 1999 radiation therapy oncology carcinoma. Cancer 89:1953–1960
group (RTOG) patients with locally advanced non-small- Zachariah B, Balducci L, Venkattaramanabalaji GV, Casey L,
cell lung cancer (LA-NSCLC): Identification of five groups Greenberg HM, DelRegato JA (1997) Radiotherapy for
with different survival. Int J Radiat Oncol Biol Phys cancer patients aged 80 and older: a study of effectiveness
48:1475–1482 and side effects. Int J Radiat Oncol Biol Phys 39:1125–1129
Radiation Therapy for Recurrent Disease
Branislav Jeremić, Jai Prakash Agarwal, and Sherif Abdel-Wahab

Contents Abstract
Recurrence is still a dominating and bitter event
1 Introduction.............................................................. 543 after the treatment of lung cancer regardless of
2 Surgery...................................................................... 545 histology, stage, or initial treatment. All recurrences
can be broadly divided into the local (e.g., lung
3 External Beam Radiation Therapy
in Locoregional Post-Surgical Recurrences parenchyma, bronchial stump, or chest wall),
of Non-Small-Cell Lung Cancer ............................ 545 regional (e.g., mediastinal lymph nodes), and distant
(e.g., brain, liver, or bones). Recurrent disease
4 External Beam Radiation Therapy
in Reirradiation of Locally Recurrent Lung almost always present as fatal event and only
Cancer ....................................................................... 546 anecdotal reports indicated potential for a treatment
5 Radiation Therapy in Reirradiation of Small-Cell to lead to cure. Beside surgery and chemotherapy,
Lung Cancer............................................................. 556 radiation therapy has been used in this setting and
6 Endobronchial Brachytherapy in Reirradiation
was shown that it was safe and effective way of
of Locally Recurrent Lung Cancer ....................... 556 retreating recurrent non-small-cell lung cancer after
initial radiation therapy. It achieved symptom
References.......................................................................... 557
control in substantial proportion of cases (overall,
approximately 80%). In many studies asymptomatic
patients were also included, indicating therefore,
intention of more ‘‘radical’’ approach of investiga-
tors. This had led to longer survivals, especially
when high-doses were successfully delivered, lead-
ing to the median survival time of 15 months and
2-year survival of 51%, not different from those
B. Jeremić (&) achieved with contemporary radiochemotherapy
Institute of Lung Diseases, Institutski Put 4, studies in treatment-naive locally advanced non-
21204 Sremska Kamenica, Serbia small-cell lung cancer. These results were obtained
e-mail: nebareje@gmail.com
at the expense of low toxicity.
B. Jeremić
Institute of Pulmonary Diseases,
Sremska Kamenica, Serbia
J. P. Agarwal
Department of Radiation Oncolgoy,
1 Introduction
Tata Memorial Center, Mumbai, India
Lung cancer remains one of the most prevalent and
S. Abdel-Wahab
Department of Oncology, Ain Shams University, deadliest malignancies worldwide. It was estimated
Cairo, Egypt that in the US in 2009 there were 219.440 new cases

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 543
DOI: 10.1007/174_2011_255, Ó Springer-Verlag Berlin Heidelberg 2011
544 B. Jeremić et al.

and estimated 159.390 deaths of lung cancer (Jemal and distant (e.g., brain, liver, or bones). Again, any
et al. 2009), making it, therefore, the major cancer combination of these may occur in a patient. Recurrent
killer in both sexes. The similar trend is seen on disease almost always present as fatal event and only
global level. The most recent data from the Interna- anecdotal reports indicated potential for a treatment to
tional Agency for the Research of Cancer (IARC) lead to cure. In addition, recurrence can also bring
showed that in 2008 worldwide there was an esti- substantial symptoms which request for additional
mated total of 1.6 millions of lung cancer cases (1.095 supportive treatment. An important issue to be
million in men and 0.514 million in women) with emphasized here is that the quality of life of such
estimated 1.38 millions of deaths (0.95 million in men patients is decreased and cost-effectiveness of diagno-
and 0.43 million in women) (IARC 2010). As such, it sis and/or treatment is always low.
represents a huge burden and big challenge for health Since some recurrences may appear in lung
care systems worldwide, which requests an orches- parenchyma (ipsilateral or contralateral lung) alone, it
trated approach in understanding and planning as to is important to recognize the distinct features of the
enable successful prevention, early diagnosis, and second metachronous primary lung cancer as opposed
treatment. to lung parenchyma recurrence occurring after the
In an attempt to optimize prevention and various initial treatment (Martini and Melamed 1975).
diagnostic and treatment approaches substantial Once recurrent lung cancer has been diagnosed,
efforts were made in combating this disease. Recent the dilemma one faces is to (re)treat or not? It is quite
decades witnessed introduction of novel molecular clear that nowadays this question should be put aside
oncology approaches as well as positron emission due to imperative in prolongation of patient’s life, and
tomography, extensively used nowadays to diagnose maintaining or improving quality of remaining life,
and stage lung cancer patients. It has also been used to unless other factors (existing serious comorbidities,
optimize radiotherapy treatment planning. In the short anticipated remaining life, and patient’s wish)
treatment domain, video assisted thoracoscopic sur- reject such an idea. Aggressive approach has support
gery, novel high-precision radiotherapy techniques in studies which showed that active treatment offers
(three-dimensional conformal radiation therapy, better outcome than purely supportive one (Hung
intensity modulated radiation therapy, stereotactic et al. 2009). Next issue to address is how to treat: with
radiation therapy), and novel chemotherapeutic agents curative or palliative intent? Here, decision-making
(third generation drugs, targeted drugs) have also process needs to be put into the context of not only
been used with increasing frequency, and, impor- the disease- and patient-related factors, but also the
tantly, efficacy. Although became more effective in level of evidence which currently exists. The data
diagnosing and treating lung cancer than ever, dismal available are from quite a few studies, all but one
and frustrating results remain even nowadays. While being retrospective and with small number of sub-
only a minority of lung cancer patients are those jects, frequently without investigation of prognostic
falling into an early stage group, where surgery is factors which may have contributed to better under-
offering hopes of cure, the vast majority of non-small- standing of both natural course of the disease and the
cell lung cancer patients and almost all with small- underlying reasons behind treatment choice. While
cell lung cancer are considered ineligible for surgery. palliation was always specified as the major aim of
There, radiation therapy and/or chemotherapy are reirradiation of locally recurrent non-small-cell lung
practiced in institutions all over the world. cancer, recent studies also included somewhat higher
In spite of promising novel biological and techno- percentage of asymptomatic patients and with wide
logical opportunities in diagnosis and treatment, introduction of novel technological appliances
recurrence is still a dominating and bitter event after the enabled higher dose to be given to more conformal
treatment of lung cancer regardless of histology, stage, volumes, leading perhaps to improved therapeutic
or initial treatment. While some failures are reported to benefit in these cases. Hence, palliative versus cura-
appear soon after the initial treatment, some manifest tive intention is likely to be revisited, especially in
years later. All recurrences can be broadly divided into cases with isolated local recurrence of previously
the local (e.g., lung parenchyma, bronchial stump, or irradiated non-small-cell lung cancer suitable for
chest wall), regional (e.g., mediastinal lymph nodes), more radical approach.
Radiation Therapy for Recurrent Disease 545

survival was 30 months and estimated 3-year survival


2 Surgery rate was 47%. Also, two recent case reports (Suzuki
et al. 2007; Neri et al. 2009) showed that lobectomy
As recurrences are documented after any treatment may provide cancer-free survivals in excess of
modality used in lung cancer they can also be treated 12 months after initial radiotherapy for stage I non-
by any of these as well. In cases of surgery small-cell lung cancer using either three-dimensional
(re-operation) in the treatment for lung cancer recur- or stereotactic radiation therapy.
rence, major determining factors were previous
complete resection, absence of other recurrent sites,
as well as the absence of concomitant diseases. Not to 3 External Beam Radiation Therapy
be forgotten, site and stage of recurrent tumor were in Locoregional Post-Surgical
considered important in decision-making process. Recurrences of Non-Small-Cell
When several large surgical series were together Lung Cancer
taken into account, it was observed that in more than
6,000 patients, mostly having intrapulmonary recur- Radiation therapy in locoregional post-surgical
rence, reoperation with curative intent was managed recurrences of non-small-cell lung cancer was used to
in 1–1.7% of patients (Gabler and Liebig 1980; treat various local/regional recurrences located intra-
Dartevelle and Khalif 1985; Watanabe et al. 1992; thoracically. Common approach was to divide these
Voltolini et al. 2000). Not unexpectedly, results were into chest wall/pleural, parenchymal, bronchial
disappointingly poor with 2-year survival of 23% stump, and mediastinal lymph node recurrences and it
(Pairolero et al. 1984) and the median survival times also could include any combination of these. Radia-
ranging from 7 to 26 months (Becker et al. 1990; tion therapy was shown to be effective and low toxic
Lesser et al. 1997; Voltolini et al. 2000; Westeel et al. in this setting (Green and Kern 1978; Kopelson and
2000). More promising results, however, were Choi 1980; Law et al. 1982; Shaw et al. 1992; Curran
observed in the most recent studies (5-year survival, et al. 1992; Yano et al. 1994; Leung et al. 1995;
15.5%), although in smaller patient population Emami et al. 1997; Kagami et al. 1998; Kono et al.
(Voltolini et al. 2000). To further extend this, even 1998; Jeremic et al. 1999; Kelsey et al. 2006).
higher local control and overall survival rates can be Observation coming from these studies is that there is
achieved by completion pneumonectomy, with 5-year dose-response favoring higher doses. Another obser-
survival of about 50% in stage I and 40% in stage II vation was that location of recurrence may influence
carcinoma (Regnard et al. 1999). Hung et al. (2009) treatment outcome, bronchial stump recurrences far-
also reported on 1- and 2-year post-recurrence sur- ing much better than recurrences located in chest
vival of 48.7 and 17.6%, respectively in patients with wall/pleura or mediastinal lymph nodes. When the
initial stage I treated surgically. Although patients data from the literature on isolated bronchial stump
who underwent complete resection for local only cases was pooled together (Jeremic and Bamberg
recurrence survived longer than those who received 2002), the median survival time was estimated to be
chemotherapy and/or radiotherapy and those who approximately 28.5 months and a 5-year survival to
received no therapy, post-recurrence survival in be about 31.5%. These results had clearly established
patients with local only recurrence was not signifi- external beam radiation therapy as a treatment of
cantly different from those with both local and distant choice in this patient population. To extend this, in a
recurrences. Newer strategies, including alternative small subset of ‘‘early’’ (i.e., stage I: T2N0) bronchial
treatment approaches, are clearly warranted in this stump recurrences in the study of Jeremic et al. (1999)
setting. Finally, recent years also brought qualita- an excellent survival (5-year: 57%) with high-dose
tively new evidence that surgery for recurrence external beam radiation therapy (C60 Gy) was
occurring after previous radiochemotherapy or achieved, which approached those obtainable with
after highly-focused radiotherapy may be beneficial. surgery alone in newly diagnosed non-small-cell lung
Bauman et al. (2008) reported on 24 patients under- cancer of the same stage (Mountain 1986; Naruke
going previous radiotherapy, 22 of whom received et al. 1998). An intriguing fact, still unexplained
concurrent chemotherapy. The median overall properly, is that their survival seems superior to that
546 B. Jeremić et al.

of patients with newly diagnosed non-small-cell lung for 5-year progression-free survival (p = 0.027). This
cancer of a similar stage when treated with high-dose study have shown that patients with postresection
standard or hyperfractionated radiation therapy (Ono recurrent non-small-cell lung cancer achieved sur-
et al. 1991; Morita et al. 1997; Jeremic et al. 1997; vival comparable to that of newly diagnosed non-
Sibley et al. 1998; Hayakawa et al. 1999; Jeremic small-cell lung cancer patients when they were both
et al. 1999). Law et al. (1982) also provided the data treated with radiation therapy or radiochemotherapy.
on patients having ‘‘more extensive’’ bronchial or These findings also suggested that patients with pos-
tracheal component of the disease. Their findings tresection recurrent chemo should be treated as
further support the effectiveness of external beam aggressively as those with newly diagnosed disease.
radiation therapy in bronchial stump recurrence.
These patients achieved the median survival time of
19 months and 1- and 3-year survival of 75, and 4 External Beam Radiation Therapy
12.5%, respectively, showing that more extensive, but in Reirradiation of Locally
ultimately localized (no nodes or other recurrent Recurrent Lung Cancer
tumor component present) may also benefit from
radiation therapy. When, however, tumor recurrence For patients recurring within previous radiation ther-
located at bronchial stump was accompanied with apy treatment field after initial treatment for either
other intrathoracic sites, such as nodal, inferior sur- early or locally advanced non-small-cell lung cancer,
vival was unequivocally documented (Curran et al. reirradiation with external beam radiation therapy was
1992; Jeremic et al. 1999; Kagami et al. 1998; Kono occasionally used. Radiation therapy was offered
et al. 1998). Contrary to these findings, Kelsey et al. aiming both increased survival and symptom control,
(2006) found superior survival for patients having with expected improvement of the quality of
mediastinal recurrence. In their study on 29 patients, remaining life.
the median radiation therapy dose was 66 Gy (range, It seems that currently there are only eleven studies
46–74 Gy). Elective nodal irradiation was used in clearly aiming to document the outcome of external
27/29 patients, while chemotherapy was given in beam radiation therapy in reirradiation of locally
15/29 patients. In an interesting study, Cai et al. recurrent non-small-cell lung cancer (Green and
(2010) have recently compared the survival of Melbye 1982; Jackson and Ball 1987; Montebello
postresection recurrent versus newly diagnosed non- et al. 1993; Gressen et al. 2000; Okamoto et al. 2002;
small-cell lung cancer patients treated with radiation Wu et al. 2003; Kramer et al. 2004; Tada et al. 2005;
therapy or radiochemotherapy. The study population Poltninikov et al. 2005; Ebara et al. 2007; Cetingoz
consisted of 661 consecutive patients with non-small- et al. 2009). In addition, one case report was identified
cell lung cancer registered in the radiation oncology on the use of intensity modulated radiation therapy as
databases at two medical centers in the United States pure planning study (Beavis et al. 2005). One study
between 1992 and 2004. Of the 661 patients, 54 had also included several different tumor entities and did
postresection recurrent non-small-cell lung cancer not specify data for reirradiation of lung cancer using
and 607 had newly diagnosed non-small-cell lung either three-dimensional or stereotactic radiation
cancer. The distribution of relevant clinical factors therapy (Jereczek-Fossa et al. 2008). Two additional
between these two groups was similar. The median studies (Coon et al. 2008; Chang et al. 2008) included
survival time and 5-year overall survival rates were patients reirradiated with sophisticated treatment
19.8 months and 14.8% versus 12.2 months and planning and stereotactic delivery in cases of locally
11.0% for recurrent versus newly diagnosed patients, recurrent non-small-cell lung cancer, but focused on
respectively (p = 0.037). For Stage I–III patients, no different aspects of radiation therapy and failed to
significant difference was observed in the 5-year produce some important patient, tumor and/or out-
overall survival (p = 0.297) or progression-free sur- come data which could have been meaningfully
vival (p = 0.935) between recurrent and newly included and compared with other studies.
diagnosed patients. For the 46 patients with Stage It is, however, virtually unknown which proportion
I–III recurrent disease, multivariate analysis showed of patients initially treated with chest radiation ther-
that chemotherapy was a significant prognostic factor apy underwent reirradiation during the natural course
Radiation Therapy for Recurrent Disease 547

of the disease. One study that addressed the issue (Table 1), five studies did not provide data on initial
(Estall et al. 2007) examined the proportion of staging (Green and Melbye, 1982; Jackson and Ball
patients who received more than one radiation therapy 1987: Gressen et al. 2000; Kramer et al. 2004; Ebara
treatment for lung cancer. While the rate of initial et al. 2007), only three (Montebello et al. 1993;
radiation therapy utilization has been estimated to be Okamoto et al. 2002; Kramer et al. 2004) provided
76% (Delaney et al. 2003) accounting for the first data on initial performance status, and only two
radiation therapy episode delivered, in the study of studies (Okamoto et al. 2002; Tada et al. 2005) pro-
Estall et al. (2007) it was 52%. Local disease in the vided data on recurrent tumor staging, while five
chest was treated with initial radiation therapy in most studies did not provide data on performance status at
cases (79%), while the second and third radiation the time of reirradiation (Green and Melbye 1982;
therapy treatment was administered in 22 and 21%, Jackson and Ball 1987: Gressen et al. 2000; Kramer
respectively. With an increase in the number of et al. 2004; Cetingoz et al. 2009). Five studies did not
treatment episodes, the mean duration between each provide data on percentage of asymptomatic patients
episode decreased as was the case with the total dose reirradiated (Green and Melbye 1982; Jackson and
and number of fractions. It is likely that this occurred Ball 1987; Gressen et al. 2000; Wu et al. 2003;
as reflection of progressive decline in performance Kramer et al. 2004). Of treatment characteristics,
status and poorer prognosis of patients in the end three studies did not provide data on cumulative
stage of their disease and their life. Unfortunately, the radiation therapy doses (Wu et al. 2003; Tada et al.
study findings are somewhat limited due to the fact 2005; Cetingoz et al. 2009) probably due to a variety
that it has covered the data from two years and not of doses per fraction used during reirradiation course.
prolonged periods of time. Unfortunately, no similar Only two studies used uniform radiation therapy
study exists to provide detailed information from doses for reirradiation (Kramer et al. 2004; Tada et al.
other institutions and regions. This could have been 2005). Two studies (Jackson and Ball 1987; Kramer
potential additional advantage of such an approach et al. 2004) did not provide data on field sizes and one
and worldwide verification because it is more than (Jackson and Ball 1987) did not provide data on
likely that different institutions/regions/countries chemotherapy administration. Tumor motion control
would have had both initial radiation therapy utili- was described in none of the eleven studies thor-
zation rate as well as reirradiation rate in lung cancer. oughly discussed, but only in two most recent studies
In general, there is both limited data in the avail- (Coon et al. 2008; Chang et al. 2008). Similarly,
able literature, vast majority of it being of retrospec- positron emission tomography-computerized scan-
tive nature. There is also great variety in not only ning was specified as done for the purpose of staging
diagnostic tools used for both initial radiation therapy and evaluation of response (not for treatment plan-
and reirradiation, but patient and tumor characteristics ning) in none of the eleven studies but only in two
(Table 1), treatment (radiation therapy) specifications most recent studies (Coon et al. 2008; Chang et al.
(Table 2), and treatment outcome (Table 3) among 2008), while Wu et al. (2003) used positron emission
available studies which documented majority of these tomography for evaluation of response in follow-up
important characteristics (7–16). Only three studies only. None of these studies or report tried to provide
(Green and Melbye, 1982; Jackson and Ball, 1987; radiobiological data that could perhaps help us more
Gressen et al. 2000) provided actual number of clearly compare effectiveness of studies. In spite of
patients with non-small-cell lung cancer from which the fact that all studies provided data on time intervals
reirradiated non-small-cell lung cancer cases were between initial radiation therapy and reirradiation,
drawn. As well, only three studies clearly defined only three studies (Montebello et al. 1993; Okamoto
time interval for considering reirradiation appropriate et al. 2002; Cetingoz et al. 2009) provided survival
treatment option for actual/presumed (depending on data from initial radiation therapy. It is therefore that
diagnostic tools used) locally recurrent non-small-cell we are left short of an opportunity to have better
lung cancer. Interestingly, there was only one (Wu insight into an overall success of not only reirradia-
et al. 2003) prospective phase I-II study. Finally, tion (as salvage radiation therapy) per se, but com-
many studies did not provide some important data at bined initial radiation therapy and reirradiation and
all. In particular, of patient and tumor characteristics perhaps compare it with the data obtained in patients
548 B. Jeremić et al.

Table 1 Patient and tumor characteristics


Author (year) N Sex Age range Initial tumor Histology PS at re-RT range Time interval from 1st
(M/F) (median) staging (%) (%) (median) to 2nd RT (median)
Green and 29 23/6 35–85 (57) n.s. SCC-66 n.s. 3–40 months (10)
Melbye ADC-14
(1982)
LC-14
SCLC-6
Jackson and 22 21/1 45–76 (62) n.s. SCC-50 n.s. 5.7–48.5 months (15)
Ball (1987) ADC-36
Other-14
Montebello 30 18/12 45–83 (62) I-II-23; SCC-53; KPS 40–100 (60) n.s. (12)
et al. (1993) IIIA-47 ADC-27
IIIB-30 LC-10
Other-10
Gressen et al. 23 13/10 47–87 (66) n.s. SCC-35 n.s. 3–156 months (15)
(2000) ADC-30
LC-9
Other-27
Okamoto et al. 34 29/5 38–85 (69) I-II-9 SCC-50 PS0-1 = 41% 5–87 months (23)
(2002) IIIA-29 ADC-18 PS C 2 = 59%
(PS2)
IIIB-53 LC-6
IV-9 Other-24
Wu et al. 23 21/2 43–79 (68) II-30 SCC-40 KPS 70–100 6–42 months (13)
(2003) III-70 ADC-30
SCLC-30
Kramer et al. 28 27/1 52–83 (68) n.s. All 6–72 months (17)
(2004) NSCLC
Tada et al. 19 17/2 49–84 (64) IIIA = 21 SCC-74 PS0-1 = 42% 5–60 months (16)
(2005) IIIB = 79 ADC-21 PS2-3 = 58%
LC-5
Poltinnikov 17 10/7 45–79 (66) n.s. SCC-35 KPS 60–90 (80) 2–39 months (13)
et al. (2005) ADC-59
Other-6
Ebara et al. 44 n.s. 49–86 (71) n.s. SCC-43 PS0-1 = 86% 5.8–47.2 months (12.6)
(2007) ADC-27 Other = 14%
SCLC-20
Other-10
Cetingoz et al. 38 35/3 33–80 (58) IIB = 5 SCC-61 n.s. 1–47 months (9)
(2009) IIIA = 10 ADC-13
IIIB = 84 Other-26
M male; F female: KPS Karnofsky performance status; RT radiotherapy; n.s. not stated; SCC squamous cell carcinoma;
ADC adenocarcinoma: LC large cell carcinoma; SCLC small cell lung cancer; other include at least two other histologies
Radiation Therapy for Recurrent Disease 549

Table 2 Treatment characteristics


Author Initial RT dose Re-RT dose Cumulative RT RT fields/volume RT field size CHT
Gy (median) Gy (median) dose Gy (median) cm2 (median) (%)
Green and 40–65 (53) 6–54 (35) 60–166* (82) Tumor only = 76% Average, 80 24
Melbye Uninvolved
(1982) mdstnm = 24%
Jackson and 50–61 (55) 20–30 (30) 70–90 (85) Volume encompassing the n.s. n.s.
Ball (1987) disease causing symptoms
Montebello 28–66 (60) 15–57 (30) 43–122* (88) Large fields** = 4–25 pts (96) 23
et al. (1993) Tumor = 30 pts (85)
Gressen et al. 25–66 (59) 6–40 (30) 60–101 (86) Tumor ? 1–2 cm 30–315 (81) 61
(2000)
Okamoto 30–80 (66) 10–70 (50) 56.5–150 (110) Radical = tumor only 20–238 (65) 47
et al. (2002) Palliative = affected region
Wu et al. 30–78 (66) 46–60 (51) n.s. Tumor ? 1.5–2.0 cm 42–210 (104) 100
(2003)
Kramer et al. 40–60 (n.s.) 16 (16) 56–76 (n.s.) ‘‘Limited RT’’ n.s. n.s.
(2004)
Tada et al. 50–70 (n.s.) 50–60 (50) n.s. ‘‘Limited RT’’ 30–204 (64) 6
(2005)
Poltinnikov 50–66 (52) 4–42 (32) n.s. GTV ? 5 mm 95 (30–189) 29
et al. (2005)
Ebara et al. 50–70-(60) 30–60 (40) 80–130 (102) Tumor ? 5–10 mm n.s. 57
(2007)
Cetingoz 29–67 (30) 5–30 (25) n.s. Tumor ? 1–2 cm 25–245 (89) 24
et al. (2009)
RT radiotherapy; CHT chemotherapy; n.s. not stated; mdstnm mediastinum; *includes patients reirradiated twice; **includes
various field sizes including ipsilateral hilum, contralateral hilum, mediastinum and ipsilateral supraclavicular fossa

with no reirradiation or those treated for an initial response (Green and Melbye 1982; Montebello et al.
tumor of the same histology and stage with radiation 1993; Tada et al. 2005; Cetingoz et al. 2009).
therapy or radiochemotherapy. To extend this, only Due to existing limitations outlined above, avail-
two studies (Montebello et al. 1993; Okamoto et al. able data may not be easy to interpret in order to
2002) provided data on actuarial local progression- instantly establish the efficacy and therapeutic benefit
free survival after reirradiation, while two studies of this approach. This mostly concerns radiation
provided data as median distant metastasis-free sur- therapy characteristics that one may want to use in
vival (Montebello et al. 1993), although some studies daily clinic such as the adequate time, dose, and
provided actual data on pattern of failure. Two studies fractionation pattern to achieve specific goal (cure,
did not report on detailed symptomatic (per symptom) palliation). In addition, there is also a legitimate
relief (Okamoto et al. 2002; Wu et al. 2003), while concern that reirradiation can cause significant tox-
two studies did not use overall symptomatic relief as icity, especially when previous high-dose radical
surrogate for treatment success (Wu et al. 2003; Tada radiation therapy was used. Nevertheless, feasibility
et al. 2005), and one (Okamoto et al. 2002) reported and efficacy of reirradiation were clearly documented
on only overall symptomatic relief. Only one study in several early reports on treatment of recurrent lung
(Montebello et al. 1993) provided symptom-free cancer (Green and Melbye 1982; Jackson and Ball
duration. Finally, five studies attempted to evaluate 1987; Montebello et al. 1993; Gressen et al. 2000).
survival according to potential prognostic factors such These studies were retrospective in nature with
as interval between first and second radiation therapy inherent bias including patients with post-surgical
course, performance status, age, size, location, or relapses, post-operatively irradiated patients, those
Table 3 Treatment outcomes (from reirradiation)
550

Author MST (range) OS Symptom improvement Toxicity (%)


months Hemoptysis Cough Chest pain Dyspnea Overall
(%) (%) (%) (%) (%)
Green and Melbye 5 (1–54) 1 yr = 14% 33 55 n.s. 44 48 Rib fracture-3
(1982) 5 yr = 3% pneumonitis-3
Jackson and Ball 5.4 (n.s) 1 yr = 38% 83 50 40 67 50 Myelopathy-5
(1987) 2 yr = 15%
Montebello et al. 5 (n.s.) n.s. 89 64 77 53 70 Esophagitis-20
(1993) skin-13
pneumonitis-3
Gressen et al. (2000) 4.9 (n.s.) 1 yr = 13% 100 60 80 73 72 Grade 5 (fatal)—4
Okamoto et al. (2002) 8 (n.s.) 1 yr = 43% n.s. n.s. n.s. n.s. 75 G2 pneumonitis-35
2 yr = 27% G3 pneumonitis-21
G2 esophagitis-12
G3 esophagitis-6
Wu et al. (2003) 14 (2–37) 1 yr = 59% n.s. n.s. n.s. n.s. n.s. G1-2 esophagitis-9
2 yr = 21% G1-2 pneumonitis-22
G3 late fibrosis-9

Kramer et al. (2004) 5.6 (n.s.) 1 yr = 18% 100 67 n.s. 35 71 G2 esophagitis-4


Tada et al. (2005) 7.1 (n.s.) 1 yr = 26% n.s. n.s. 80 100 n.s. G3 pneumonitis-5
2 yr = 11% G2 esophagitis-16
Poltinnikov et al. 5.5 (2.5–30) n.s. n.s. n.s. n.s. 65 G1 esophagitis-18
(2005) G2 esophagitis-24
G1 pneumonitis-6
Ebara et al. (2007) 6.5 1 yr = 27.7% n.s. n.s. n.s. 81 G2 pneumonitis-7
G3 pneumonitis-7
Cetingoz et al. (2009) 3 (n.s.) 1 yr = 8.7% 86 77 60 69 78 G1-2 esophagitis-77
2 yr = 5.8% G3 esophagitis-4
MST median survival time; OS overall survival; n.s. not stated; G grade
B. Jeremić et al.
Radiation Therapy for Recurrent Disease 551

with metastasis and those with second primary lung beam radiation therapy through a prospective phase
cancer. Doses of the initial course of radiation therapy I–II study. After reirradiation, the median survival
ranged from 25 to 66 Gy, while those administered at time was 14 months and a 2-year survival rate was
the time of recurrence ranged from 6 to 57 Gy. 21%, while 2-year locoregional progression-free sur-
Therefore, cumulative doses ranged from 43 to vival was 42%. No acute grade C3 toxicity had been
122 Gy. Few patients underwent even third course of reported during the follow up (median 15 months
radiation therapy (second reirradiation). Contrary to after the end of reirradiation). Seventeen (74%)
radiation therapy treatment fields used during the patients had either grade 0 or 1 late toxicity. Six
initial course of radiation therapy, which were usually (26%) patients had pulmonary fibrosis on computer-
including more or less of uninvolved (prophylactic) ized tomography scan, of which symptomatic
nodal regions, those used at the time of reirradiation (grade 3) was observed in 2 (9%) patients. Besides
were obviously limited, in general only including promising results, this study also showed that its
visible recurrence with a safety margin of 1–2 cm prospective character may have enabled better
(Green and Melbye 1982; Jackson and Ball 1987; documentation of not only acute, but also late toxic-
Montebello et al. 1993; Gressen et al. 2000). Fear of ity. This may be especially important with expected
excessive toxicity, which primarily may have occur- better results, including longer survivals newer tech-
red in lung and spinal cord, clearly governed the nological tools may bring with successful dose esca-
choice of both total dose and treatment field used lation owing to better imaging and dose conformation
during the reirradiation. Symptom relief was the main deemed necessary for more curative approaches.
goal of reirradiation. In a report of Gressen et al. The prospective study of Kramer et al. (2004)
(2000), clinical data of existing articles were sum- confirmed this observation, using 2 fractions of 8 Gy
marized indicating a control of hemoptysis in 83%, given with one week split, a practical and comfortable
cough in 65%, dyspnoea in 60%, and pain in 64% of palliative regimen for both patients and hospitals. The
cases. Reirradiation seemed to be less hazardous than median survival time of 5.6 months was accompanied
anticipated with a merely 3–5% complication rate with Karnofsky performance status score being
(Green and Melbye 1982; Jackson and Ball 1987; improved in 45% patients. The overall median dura-
Montebello et al. 1993; Gressen et al. 2000). tion of symptom relief was 4 months.
The last decade brought several studies which Contrary to Kramer et al. (2004), Tada et al. (2005)
provided additional evidence on effectiveness and low used more radical approach with curative intent. They
toxicity of external beam radiation therapy in the have treated 19 patients with stage III non-small-cell
treatment of locally recurrent non-small-cell lung lung cancer with 50 Gy in 25 daily fractions,
cancer. Okamoto et al. (2002) reported on 29 males including one patient treated with 60 Gy in 30 daily
and 5 females with initially irradiated stage I–IV non- fractions. Five patients could not receive the pre-
small-cell lung cancer that have been reirradiated scribed reirradiation dose. Overall response rate was
after a median time of 23 months (range, 43%. The overall 1-year and 2-year survival rates
5–87 months). Median initial radiation therapy dose were 26 and 11%, respectively, and the median sur-
of 66 Gy (range, 30–80 Gy) was followed by median vival time was 7.1 months.
reirradiation dose of 50 Gy (range, 10–70 Gy), lead- Ebara et al. (2007) reported on a study with clearly
ing to a median of 110 Gy total cumulative dose defined palliation as a goal. In the largest series
(range 56.5–110 Gy). Authors clearly divided their published so far, the authors included 44 patients with
treatment into a radical and palliative, which affected in-field recurrent lung cancer after definitive radiation
their treatment field design choice. Calculating from therapy. Symptom relief was assessed according to
the start of the initial radiation therapy, the median scoring system created by Kramer et al. (2004). While
survival time was 31 months and 2- and 5-year initial radiation therapy doses ranged 50–70 Gy those
overall survival rates were 65% and 33%, of reirradiation were 30–40 Gy. Chemotherapy was
respectively. initially given in 25 (57%) cases and at the time of
The study of Wu et al. (2003) was the first ever to reirradiation in 16 (36%) cases. Size of radiation
address the issue of reirradiation of 23 locoregionally therapy fields used at initial radiation therapy ranged
recurrent lung cancer patients after previous external 26–288 cm2 (median, 104 cm2), while those of
552 B. Jeremić et al.

reirradiation ranged 16–100 cm2 (median, 48 cm2). a significant advantage (e.g., shape and location of the
Symptom relief was observed in 74% of patients. The tumor as well as in cases of reirradiation), it is
median survival time was 6.5 months and 1-year expected that this technique plays an important role in
survival was 27.7% with only 3% patients experi- reirradiation of lung cancer in the future.
encing Grade 3 pneumonitis. Distant metastasis at Poltinnikov et al. (2005) were the first to report on
reirradiation influenced survival favoring patients the use of hypofractionated stereotactic fractionated
without it (1- and 2-year rates, 31.3% and 31.3% radiation therapy in 17 patients previously irradiated
versus 25% and 0%, respectively; p = 0.02). Of all to a median dose of 52 Gy (range, 50–66 Gy) deliv-
variables investigated upon influence on survival, ered with concurrent chemotherapy in all cases. The
only additional chemotherapy achieved statistical median time interval between completion of the first
trend toward improved survival in both univariate course of radiation therapy and the start of reirradia-
(p = 0.067) and multivariate (p = 0.090) model. tion was 13 months (range, 2–39 months). A median
The most recent report on the use of external beam dose of hypofractionated schedule was 32 Gy (range,
radiation therapy in reirradiation in non-small-cell 4–42 Gy), with a median fraction size of 4 Gy
lung cancer is that of Cetingoz et al. (2009) who (range, 2.5–4.2 Gy) delivered 3–5 times per week.
reported the study comprising of 38 patients. Most of Five patients also received concurrent chemotherapy.
the patients (81%) were previously treated with pal- Radiologic response was observed in 5 (29%),
liative intent due to poor prognostic factors such as and stable disease in another 5 (29%) patients. The
advanced disease, poor performance status and pro- median survival was from the start of reirradiation
nounced weight loss. Initial radiation therapy doses was 5.5 months (range, 2.5–30 months). Symptom
ranged 28.8–67.2 Gy (median, 30 Gy) in 9–33 resolution was observed in 11/13 (85%) symptomatic
fractions (median, 10 fractions) with 2–3.2 Gy per patients, while no change was observed in the
fraction. During reirradiation, a median total dose of remaining two (15%) patients. No Grade 3 or higher
25 Gy (range, 5–30 Gy) in median of 10 fractions side-effects were observed.
(range, 1–10 fractions) with median of 3 Gy (range, Three additional reports on the use of stereotactic
2–10 Gy) per fraction were delivered. The median radiation therapy in recurrent non-small-cell lung
overall survival time (from the first irradiation) was cancer were recently published. Chang et al. (2008)
13.5 months (range, 4–65 months) and the median used four-dimensional computerized tomography-
survival time after reirradiation was 3 months (range, based planning to deliver 40–50 Gy to 14 patients
0–65 months). One-year and two-year survival were with isolated recurrent non-small-cell lung cancer
57.8 and 28.8%, respectively after initial diagnosis. previously treated with definitive radiation therapy
Recent years brought first reports on the use of with or without chemotherapy or surgical resection
highly sophisticated radiation therapy planning and before stereotactic radiation therapy. With a median
execution in cases of reirradiation of mostly non- follow-up of 17 months (range, 6–40 months), the
small-cell lung cancer, such as stereotactic radiation crude local control at the treated site was 100% for
therapy or intensity modulated radiation therapy patients treated using 50 Gy. Four (29%) patients
(Beavis et al. 2005; Jereczek-Fosasa et al. 2008; Coon developed Grade 2 pneumonitis, but no patient
et al. 2008; Chang et al. 2008). Beavis et al. (2005) experienced grade C3 toxicity. Coon et al. (2008)
provided first report of the planning study on the use reported on a similar fractionated stereotactic radia-
of another novel radiation therapy technique, intensity tion therapy but using novel technological tool called
modulated radiation therapy, in the re-treatment of a Cyber Knife. For all (n = 12) patients with recurrent
patient with non-small-cell lung cancer. Although tumors, a dose of 60 Gy was given in 3 fractions.
similar distributions were achieved for organs at risk Importantly, in majority of patients pre-treatment
by intensity modulated radiation therapy versus con- positron emission tomography-computerized tomog-
ventional technique, the target coverage given by the raphy scans were performed to aid in delineation of
conventional treatment option was clearly inferior to tumor volume. Also, all patients received regularly
that offered by the intensity modulated radiation scheduled follow-up planned computerized tomogra-
therapy plan. With the widespread use of intensity phy or positron emission tomography-computerized
modulated radiation therapy in cases when it can be of tomography imaging. Overall response rate was 75%,
Radiation Therapy for Recurrent Disease 553

while 17% of patients experienced stable disease. At a 8 months and a 2-year survival was 27%, for the
median follow-up of 11 months, the local control rate patients treated with curative intent and higher radi-
at the site treated was 92% and overall survival was ation therapy doses, the median survival time was as
67%. Only one (2%) patient experienced Grade 2 high as 15 months and 2-year survival was 51%.
pneumonitis. Most recently, Kelly et al. (2010) ret- Contrasting this is the observation of Jackson and Ball
rospectively reviewed outcomes after stereotactic (1987) who did not find difference between patients
radiation therapy for recurrent disease among patients retreated with 20 Gy and those retreated with 30 Gy,
previously given radiation therapy to the chest at The possibly because both dose levels were relatively low.
MD Anderson Cancer Center. There were 36 such Symptom relief varied between studies and
cases and the median follow-up time after stereotactic between symptoms investigated. Hemoptysis was
radiation therapy was 15 months. Stereotactic radia- controlled in 33–100% cases, but when study of
tion therapy provided in-field local control for 92% of Green and Melbye (1982) was excluded due to small
patients; at 2 years, the actuarial overall survival rate number of events (1 out of 3 responders), it became
was 59%, and the actuarial progression-free survival rather uniform (83–100%). Cough was controlled in
rate was 26%, with the primary site of failure being 43–77% cases, chest pain in 40–80% cases, while
intrathoracic relapse. Fifty percent of patients expe- dyspnea in 35–100% cases, the latter event having
rienced worsening of dyspnea after stereotactic greater range possibly again due to somewhat small
radiation therapy, with 19% requiring oxygen sup- number of events reported in some studies. When,
plementation; 30% of patients experienced chest wall however, overall symptom relief was considered,
pain and 8% grade 3 esophagitis. No grade 4 or 5 except aforementioned studies of Green and Melbye
toxic effects were noted. This study confirmed pre- (1982) (48%) and Jackson and Ball (1987) (50%), it
vious, though limited, observations that stereotactic became rather uniform, ranging 70–78% cases.
radiation therapy can provide excellent in-field tumor Reirradiation seemed to be less hazardous than
control in patients who have received prior radiation anticipated with a merely 5% complication rate
therapy. Toxicity was significant but manageable. The observed in initial studies (Green and Melbye, 1982;
high rate of intrathoracic failure in this study indi- Jackson and Ball, 1987; Montebello et al. 1993). The
cated the need for further study to identify patients most frequent event was esophagitis (20%), but
who would derive the most benefit from stereotactic pneumonitis appeared in only 3% of cases, with
radiation therapy in this setting. myelopathy and rib fracture being a rare and anec-
When overall results of external beam radiation dotal event. These data may have suffered from both
therapy in reirradiation of locally recurrent lung underreporting and the lack of detailed scoring sys-
cancer using different endpoints of studies are sum- tems used in the past. When however, more recent
marized, the median survival times ranged from 3 to studies using scoring systems are taken into account,
14 months, while 1- and 2-year survival rates ranged grade 3 esophagitis was noted in only 4–6% cases,
from 8.7–59 and from 5.8–27%, respectively. In and grade 3 pneumonitis was noted in 5–21% cases.
the series of Jackson and Ball (1987), 4-year rate Somewhat higher incidence (21%) occurring in the
was 15 months. Interestingly, the three studies study of Okamoto et al. (2002) may have been the
that provided the longest median survival time consequence of the highest total cumulative and
(7.1–14 months) and highest survival rates (Okamoto highest median cumulative doses (150 and 110 Gy,
et al. 2002; Wu et al. 2003; Tada et al. 2005) all used respectively) used. Additional support about possible
reirradiation doses with median values of 50–51 Gy, dose-effect for such an observation lies in the fact that
which was in contrast to studies using lower reirra- grade 2 pneumonitis occurred in that study in 35%
diation doses which achieved the median survival cases, while in other studies it was much less. Similar
time of 4.9–5.6 months, indicating possible dose- total doses of initial radiation therapy and reirradia-
effect. Furthermore, in the study of Tada et al. (2005) tion may have been the cause of grade 2–3 late lung
the median survival time for 14 patients who received fibrosis occurring in 26% cases in the study of Wu
the prescribed dose was 10.5 months. Similarly, in the et al. (2003), of which only 2 (9%) was Grade 3. One
study of Okamoto et al. (2002), while for the whole should not forget that, contrary to other studies which
group of patients, the median survival time was largely lacked detailed late toxicity data, this study
554 B. Jeremić et al.

was the only prospective study which have enabled study to influence survival (9), with patients with
prospective data collection and therefore, better doc- stage I and II having better survival then those with
umentation of the events, especially important for late IIIA or IIIB, with the latter two groups of patients
toxicity. Toxicity data also reconfirmed that occa- having similar survival. Also, an improvement in
sional fatal (grade 5) event may occur. Although in symptom (dyspnea) control did not lead to improved
the study of Gressen et al. (10) a single patient died survival in the study of Gressen et al. (2000).
due to possible radiation therapy pneumonitis author General observation coming from the literature is
did not exclude a possibility of the disease progres- that external beam radiation therapy is safe and
sion after receiving 30 Gy of reirradiation together effective way of retreating recurrent non-small-cell
with concurrent and post-radiochemotherapy. lung cancer after initial radiation therapy. It achieved
Of all potential prognostic factors investigated, age symptom control in substantial proportion of cases
was not shown to influence survival (Gressen et al. (overall, approximately 80%). In many studies
2000; Ebara et al. 2007; Cetingoz et al. 2009). asymptomatic patients were also included, indicating
Additionally, no difference in the treatment outcome therefore, intention of more ‘‘radical’’ approach of
between patients \70 years and those C70 years was investigators. This had led to longer survivals, espe-
observed (Gressen et al. 2000; Ebara et al. 2007). This cially when high-doses were successfully delivered,
may indicate greater applicability of external beam leading to the median survival time of 15 months and
radiation therapy in this disease in elderly, in partic- 2-year survival of 51%, not different from those
ular, irrespective of treatment intention and also when achieved with contemporary radiochemotherapy
severe late effects may become less important. Also, studies in treatment-naive locally advanced non-
neither tumor size nor its location was shown to small-cell lung cancer. They were also accompanied
influence outcome (Cetingoz et al. 2009). Treatment with very low high grade toxicity (3–5%), especially
response may have positively influenced treatment when modern radiation therapy technologies were
outcome (Green and Melbye 1982) as well as good used. It seems that a course of both palliative radia-
performance status (Green and Melbye 1982; Tada tion therapy (e.g., 30 Gy in 10 daily fractions) or
et al. 2005), but not unequivocally (Ebara et al. 2007). more radical radiation therapy (e.g., 50 Gy in 25 daily
In the study of Tada et al. (2005), performance status fractions) can be either repeated after initial radiation
influenced survival being 12.6, 7.1, and 1.1 months therapy leading to total cumulative doses of
for patients with performance status 0–1, performance 85–100 Gy without increased toxicity after a time
status 2, and performance status 3, respectively. The interval of 10–12 months. While no formal radiobi-
variable that was shown that may influence treatment ological modeling was ever attempted to bring more
outcome was time interval between the end of first insight into ‘‘recommended’’ dose(s), these ‘‘obser-
radiation therapy and reirradiation. Interestingly, vations’’ from the data from the literature should be
studies of Tada et al. (2005) and Cetingoz et al. used cautiously.
(2009) showed that longer intervals may be advanta- Although there is great variety of radiation therapy
geous for better survival. In addition to it, Kramer characteristics, especially total dose, dose per fraction
et al. (2004) observed that none of the patients with an and dose prescriptions used, likely as the cause of
interval \10 months survived [5 months, while all different techniques used, additional observations and
patients surviving [1 year had a tumor-free interval conclusions would include the following:
of [1 year. Contrary to that, in the study of Gressen (1) there is tendency toward the use of smaller mar-
et al. (2000) the time interval (C10 months ver- gins during treatment planning, especially in
sus \10 months), did not influence outcome, simi- more recent studies, using sophisticated tech-
larly to the study of Ebara et al. (2007) who could not niques of treatment planning; they allow high-
document it when cut-off value was 13 months. Dose- precision of dose delivery to as conformal vol-
response was evaluated in the study of Jackson and umes as possible as necessary mode of achieving
Ball (1987) and was found similar between 20 and total cumulative doses that surpass known total
30 Gy (median doses). No influence of pre-radiation tolerance doses in cases of primary, treatment-
therapy and reirradiation dose was found in the study naive non-small-cell lung cancer (Emami et al.
of Ebara et al. (2007). Stage was also found in one 1991),
Radiation Therapy for Recurrent Disease 555

(2) there is tendency for better reporting of toxicity the median survival time associated with time
occurring during and after reirradiation using intervals of less than 12 months, 12–18 months
toxicity scoring systems; this is especially and more than 18 months were 2.1, 7.1, and
important due to a need for better reporting of late 11.5 months, respectively. Although Gressen
toxicity, an additional advantage for clinicians et al. (2000) did not observe this influence, this
and radiobiologists when large total cumulative was recently reconfirmed by Cetingoz et al.
doses are concerned, (2009) who performed a multivariate analysis to
(3) there is a general lack of radiobiological impact show that time interval between the first radiation
in this field, a disappointing fact due to some- therapy and reirradiation was the only indepen-
times surprisingly high total cumulative doses dent prognosticator influencing overall survival.
which exceeded general belief of normal tissue These findings may imply less aggressive
tolerance (Emami et al. 1991), especially when behavior of tumors being both diagnosed and
this was coupled with low toxicity, reirradiated later, or simply better local control
(4) Even in cases when chemotherapy had been achieved in cases of reirradiation occurring later,
administered initially, concurrent chemotherapy but also preference of involved radiation oncol-
seems feasible to be safely administered during ogists to use higher doses with prolonged time
reirradiation course due to low toxicity in studies intervals between first radiation therapy and re-
reporting its use, and irradiation. Although not specified in any study,
(5) time intervals between the initial radiation ther- the latter item need to be put into the context of
apy and reirradiation were specified more fre- speculation whether likelihood of reirradiation
quently in more recent studies (Okamoto et al. (and if so, its characteristics) was affected by
2002; Wu et al. 2003; Tada et al. 2005; Ebara initial radiation therapy characteristics. Interest-
et al. 2007; Cetingoz et al. 2009; Poltninikov ingly, in a large systematic review of palliative
et al. 2005), but not always (Coon et al. 2008; thoracic radiation therapy there was more reirra-
Chang et al. 2008). In particular, the issue of time diation to the thorax after low-dose palliative
interval between first and second radiation ther- radiation therapy compared to high-dose, but this
apy may be an important issue for better under- was insignificant (Fairchild et al. 2008).
standing natural history of the disease, its Also, novel technologies, diagnostic and thera-
potential influence on decision-making process, peutic, such as stereotactic radiation therapy, intensity
and discussing the issue of potential prognostic modulated radiation therapy, protons, or carbon ions
factors as well as toxicity expected to occur to a could enable earlier diagnosis as well as successful
lesser degree after prolonged time periods dose escalation with low toxicity and provide neces-
between first and second radiation therapy. Reir- sary tool for reirradiating local recurrence which need
radiation started as early as 1–6 months after the radiation therapy with or without concurrent chemo-
first radiation therapy course and was as late as therapy. Taking into account previous radiation ther-
39–87 months after it, with the similar median apy characteristics, performance status and time
values of 13–16 months (Wu et al. 2003; Tada interval between initial radiation therapy and irradi-
et al. 2005; Ebara et al. 2007; Poltninikov et al. ation could be seen as a starting point for discrimi-
2005) and only exception were studies of nating curative from palliative intent in these patients
Cetingoz et al. (2009) where it was 8.5 months with former case advocating for higher doses, smaller
and the study of Okamoto et al. (2002) where it fields, and possible administration of concurrent
was 23 months. Importance of understanding the chemotherapy. Local recurrences not suitable for
influence of time interval between the first radi- radical treatment may require palliative radiation
ation therapy and reirradiation comes from the therapy with fewer fractions given in shorter time.
study of Tada et al. (2005) who showed that While no clearly established guidelines can be
besides performance status, time interval was expected to appear soon helping clinicians in this
another important factor influencing treatment early task, due to great variety in basic tumor, patient
outcome. Interval between first and second radi- and radiation therapy characteristics of the accumu-
ation therapy also influenced treatment outcome: lated data from the literature, current wisdom calls for
556 B. Jeremić et al.

prudent use of available technology in external beam different sources such as 137-CS, 198-Au, or 192-Ir,
radiation therapy for reirradiation of locally recurrent sometimes combined with low-dose external beam
lung cancer, with clear objectives set up front. radiation therapy (Mendiondo et al. 1983) with sat-
isfactory palliative results. Subsequent studies pro-
vided evidence on the effectiveness of endobronchial
5 Radiation Therapy in Reirradiation brachytherapy using different dose rate in this disease.
of Small-Cell Lung Cancer The vast majority of these used high-dose rate
(Seagren et al. 1985; Mehta et al. 1989; Bedwinek
Radiation therapy was infrequently used to treat et al. 1991; Sutedja et al. 1992; Gauwitz et al. 1992;
locoregional recurrence of small-cell lung cancer. Gustafson et al. 1995; Micke et al. 1995; Delclos et al.
Because of the fear that it may add only toxicity 1996; Ornadel et al. 1997; Hatlevoll et al. 1999; Kelly
without clear benefit for patients, this was especially et al. 2000; Zorlu et al 2008; Hauswald et al. 2010).
in cases of limited disease previously treated with a Importantly, in majority of reports previous external
combined radiochemotherapy approach. In cases of beam radiation therapy was used with median doses
extensive disease, radiation therapy at the time of mostly ranging 54–58 Gy (Bedwinek et al. 1991;
recurrence after initial chemotherapy also can be Sutedja et al. 1992; Gauwitz et al. 1992; Gustafson
considered, but this was mostly related to a symp- et al. 1995; Micke et al. 1995; Hauswald et al. 2010),
tomatic patient. Investigators (Ihde et al. 1979; Ochs although in the study of Zorlu et al. (2008) the median
et al. 1983; Salazar et al. 1991) used the doses as low total conventional equivalent dose was 30 Gy (range,
as 21 Gy but also as high as 60 Gy in patients har- 30–70 Gy). Only rarely studies reported on the use of
boring recurrences from both limited and extensive a single fraction of endobronchial irradiation of either
disease small-cell lung cancer. Observed responses 10 Gy (Seagren et al. 1985; Hatlevoll et al. 1999;
within the radiation therapy field ranged 52–77% Zorlu et al. 2008), or 15 Gy (Zorlu et al. 2008), or
patients, with the median survival times ranging only 20–30 Gy (Mehta et al. 1989). The dose per fraction
3–4 months. Likely cause of such dismal figures ranged 6–15 Gy, while in two German studies (Micke
seems to be early systemic progression. Nevertheless, et al. 1995; Hauswald et al. 2010) it was 5 Gy,
the wide range of doses used gave an opportunity to delivered in 2–4 fractions. Subjective response to
the authors to speculate about higher doses (C40 Gy) treatment was observed in 66–94%. In the study of
producing better palliation, an important matter in Ornadel et al. (1997), the percentage of patients with
patients with limited remaining lifetime. scores 0 or 1 (none or mild) for each symptom pre-
treatment and at 3 months were as follows: cough
62–77% (43% improving by at least one grade),
6 Endobronchial Brachytherapy dyspnea 32–56% (50% improvements by at least one
in Reirradiation of Locally grade), hemoptysis 78–97%, and performance status
Recurrent Lung Cancer 65–84% (54% by at least one grade). In the study of
Zorlu et al. (2008) performance status was improved
In addition to external beam radiation therapy, and symptomatology reduced in 81% of patients. Ten
endobronchial brachytherapy was occasionally used out of 14 (71%) dyspneic patients recovered clini-
alone to treat recurrent bronchogenic carcinoma. This cally, while hemoptysis in 5 (36%) patients recov-
was especially so in cases when previous external ered. However, the mean period of palliation was
beam radiation therapy has been given. Similarly to disappointingly low, being only 45 days (range,
the literature on the use of external beam radiation 0–9 months). The period of palliation was signifi-
therapy, the vast majority of reports included various cantly longer in patients with high Karnofsky Per-
histologies combined with only a minority of patients formance Status score (C80) at the initial evaluation.
having small-cell histology. Unfortunately, some The most recent study of Hauswald et al. (2010)
reports included even patients with primary lung showed that relief of symptoms was documented as
carcinomas, additionally obscuring the picture. More excellent in 12% of patients and good in 46% of
than 25 years ago early reports provided different patients. Complete remission was observed in 15% of
aspects of endobronchial radiation therapy with patients, and partial response in 58% of patients,
Radiation Therapy for Recurrent Disease 557

while 10% of patients showed no response to treat- perspective of cost-benefit analysis, especially when
ment and 15% had progressive disease. In other single-fraction HDR is considered. Regardless of
studies, objective response measured during bron- these shortcomings, endobronchial brachytherapy
choscopy was observed in 72–100% patients, while remains to be one of the cornerstones of successful
radiologic documentation of re-aeration was observed palliative approaches in patients with symptomatic
in 64–88% patients. Duration of response ranged endobronchial recurrences of lung cancer. This
4.5–6.5 months. Actuarial local control rates were treatment modality will be dealt upon in more detail
rarely reported, being in the most recent study in another chapter.
of Hauswald et al. (2010) 17% at 1-year and 3% at
2-years, respectively. In that study, the median local
progression-free survival time was 4 months (range, References
1–23 months). Reported survivals were as high as
25% at 1 year (Bedwinek et al. 1991), while Kelly Bauman JE, Mulligan MS, Martins RG, Kurland BF, Eaton KD,
et al. (2000) and Hauswald et al. (2010) achieved Wood DE (2008) Salvage lung resection after definitive
radiation ([59 Gy) for non-small cell lung cancer: surgical
identical survival of 18% and 7% at 1- and 2-year, and oncologic outcomes. Ann Thorac Surg 86:1632–1638
respectively). The median survival time ranged Beavis AW, Abdel-Hamid A, Upadhyay S (2005) Re-retreat-
5–9 months (Bedwinek et al. 1991; Gauwitz et al. ment of a lung tumour using a simple intensity-modulated
1992; Delclos et al. 1996; Micke et al. 1995; Kelly radiotherapy approach. Br J Radiol 78:358–361
Becker HP, Radomsky J, Hartel W (1990) Das rezidiv des
et al. 2000; Zorlu et al. 2008; Hauswald et al. 2010) nicht-kleinzelligen bronchialkarzinoms: indikation und
and two studies reported identical median survival ergebnisse der chirirgischen behandlung. Penumologie
time of 7 months for responders (Sutedja et al. 1992; 44:1106–1109
Kelly et al. 2000). Various treatment-related compli- Bedwinek J, Petty A, Bruton C, Sofield J, Lee L (1991) The use
of high dose rate endobronchial brachytherapy to palliate
cations have been observed; the most feared being symptomatic endobronchial recurrence of previously irra-
fatal bleeding. Incidence of severe pulmonary bleed- diated bronchogenic carcinoma. Int J Radiat Oncol Biol
ing in initial reports (Seagren et al. 1985; Bedwinek Phys 22:23–30
et al. 1991; Sutedja et al. 1992) ranged 25–32%, while Cai XW, Xu LY, Wang L, Hayman JA, Chang AC, Pickens A,
Cease KB, Orringer MB, Kong FM (2010) Comparative
most recent studies (Gauwitz et al. 1992; Gustafson survival in patients with postresection recurrent versus
et al. 1995; Delclos et al. 1996; Kelly et al. 2000; newly diagnosed non-small-cell lung cancer treated with
Zorlu et al. 2008; Hauswald et al. 2010) reported on radiotherapy. Int J Radiat Oncol Biol Phys 76:1100–1105
significantly lower incidence of this complication Cetingoz R, Arikan-Alicikus Z, Nur-Demiral A, Durmak-Isman
B, Bakis-Altas B, Kinay M (2009) Is reirradiation effective
ranging from 0 to 7%. Although a number of factors in symptomatic local recurrence of non small cell lung
were investigated upon their influence of the inci- cancer patients? A single institution experience and review
dence of fatal bleeding, no firm conclusion could be of the literature. J BUON 14:33–40
drawn due to different nature of reporting (crude Chang JY, Balter PA, Dong L, Yang Q, Liao Z, Jeter M, Bucci
MK, McAleer MF, Mehran RJ, Roth JA, Komaki R (2008)
versus actuarial), and due to frequently lacking pre- Stereotactic body radiation therapy in centrally and supe-
treatment patient and tumor characteristics. However riorly located stage I or isolated recurrent non-small-cell
in one such attempt, Ornadel et al. (1997) identified lung cancer. Int J Radiat Oncol Biol Phys 72:967–971
prior laser resection as major factor contributing to a Coon D, Gokhale AS, Burton SA, Heron DE, Ozhasoglu C,
Christie N (2008) Fractionated stereotactic body radiation
risk (20% at 2 years in their study) of fatal hemopt- therapy in the treatment of primary, recurrent, and meta-
ysis. While majority of studies concentrated on the static lung tumors: the role of positron emission tomogra-
incidence of fatal hemoptysis, Hauswald et al. (2010) phy/computed tomography-based treatment planning. Clin
also provided detailed analysis of other side-effects of Lung Cancer 9:217–221
Curran WJ Jr, Herbert SH, Stafford PM, Sandler HM, Rosenthal
this treatment modality, such as tissue necrosis, pen- SA, McKenna WG, Hughes E, Dougherty MJ, Keller S
umothoraces causing dyspnoea, bronchomediastinal (1992) Should patients with post-resection locoregional
fistulas or mild hemoptysis not requiring transfusion. recurrence of lung cancer receive aggressive therapy? Int J
It became obvious that detailed documentation of all Radiat Oncol Biol Phys 24:25–30
Dartevelle P, Khalif J (1985) Surgical approach to local
side-effects occurring during and after the treatment is recurrence and the second primary lesion. In: Delarue NC,
necessary as to enable one to put overall results of Eschapasse H (eds) International trend in general thoracic
endobronchial brachytherapy in this setting also into a surgery, vol 1. WB Saunders, Philadelphia, pp 156–163
558 B. Jeremić et al.

Delaney GD, Jacob S, Freatherstone C (2003) A model for Jackson MA, Ball DL (1987) Palliative retreatment of locally
decision making for the use of radiotherapy in lung cancer. recurrent lung cancer after radical radiotherapy. Med J Aust
Lancet Oncol 4:120–128 147:391–394
Delclos ME, Komaki R, Morice RC, Allen PK, Davis M, Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ (2009)
Garden A (1996) Endobronchial brachytherapy with high- Cancer Statistics. CA Cancer J Clin 59:225–249
dose-rate remote afterloading for recurrent endobronchial Jereczek-Fossa BA, Kowalczyk A, D’Onofrio A (2008) Three-
lesions. Radiology 201:279–282 dimensional conformal or stereotactic reirradiation of
Ebara T, Tanio N, Etoh T, Shichi I (2007) Palliative re-irradiation recurrent, metastatic or new primary tumors. Analysis of
for in-field recurrence after definitive radiotherapy in patients 108 patients. Strahlenther Onkol 184:36–40
with primary lung cancer. Anticancer Res 27:531–534 Jeremic B, Bamberg M (2002) External beam radiation therapy
Emami B, Lyman J, Brown A (1991) Tolerance of normal for bronchial stump recurrence of non-small-cell lung cancer
tissue to therapeutic irradiation. Int J Radiat Oncol Biol after complete resection. Radiother Oncol 64:251–257
Phys 21:109–122 Jeremic B, Shibamoto Y, Acimovic LJ, Milisavljevic S (1997)
Emami B, Graham MV, Deedy M, Shapiro S, Kucik N (1997) Hyperfractionated radiotherapy alone for clinical stage I
Radiation therapy for intrathoracic recurrence of non-small nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys
cell lung cancer. Am J Clin Oncol (CCT) 20:46–50 38:521–525
Estall V, Barton MB, Vinod SK (2007) Patterns of radiotherapy Jeremic B, Shibamoto Y, Milicic B, Milisavljevic S, Nikolic N,
re-treatment in patients with lung cancer: a retrospective, Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999)
longitudinal study. J Thorac Oncol 2:531–536 External beam radiation therapy alone for loco-regional
Fairchild A, Harris K, Barnes E (2008) Palliative thoracic recurrence of non-small-cell lung cancer after complete
radiotherapy for lung cancer. J Clin Oncol 26:4001–4011 resection. Lung Cancer 23:135–142
Gabler A, Liebig S (1980) Reoperation for bronchial carci- Kagami Y, Nishio M, Narimatsu N, Mjoujin M, Sakurai T,
noma. Thorax 35:668–674 Hareyama M, Saito A (1998) Radiotherapy for locoregional
Gauwitz M, Ellerbroek N, Komaki R, Putnam JB Jr, Ryan MB, recurrent tumours after resection of non-small cell lung
DeCaro L, Davis M, Cundiff J (1992) High dose endobron- cancer. Lung Cancer 20:31–35
chial irradiation in recurrent bronchogenic carcinoma. Int J Kelly JF, Delclos ME, Morice RC, Huaringa A, Allen PK,
Radiat Oncol Biol Phys 23:397–400 Komaki R (2000) High-dose-rate endobronchial brachyther-
Green N, Kern W (1978) The clinical course and treatment apy effectively palliates symptoms due to airway tumours: the
results of patients with postresection locally recurrent lung 10-year M. D. Anderson cancer center experience. Int J
cancer. Cancer 42:2478–2482 Radiat Oncol Biol Phys 48:697–702
Green N, Melbye RW (1982) Lung cancer: retreatment of local Kelly P, Balter PA, Rebueno N, Sharp HJ, Liao Z, Komaki R,
recurrence after definitive irradiation. Cancer 49:865–868 Chang JY (2010) Stereotactic body radiation therapy for
Gressen EL, Werne-Wasik M, Cohn J, Topham A, Curran WJ patients with lung cancer previously treated with thoracic
Jr (2000) Thoracic reirradiation for symptomatic relief after radiation. Int J Radiat Oncol Biol Phys 78:1387–1393
prior radiotherapeutic management for lung cancer. Am J Kelsey CR, Clough RW, Marks LB (2006) Local recurrence
Clin Oncol (CCT) 23:160–163 following initial resection of NSCLC: salvage is possible
Gustafson G, Vicini F, Freedman L, Johnston E, Edmudson G, with radiation therapy. Cancer J 12:283–288
Sherman S, Pursel S, Komic M, Chen P, Borrego JC, Kono K, Murakami M, Sasaki R (1998) Radiation therapy for
Seidman J, Martinez A (1995) High dose rate endobronchial non-small cell lung cancer with postoperative intrathoracic
brachytherapy in the management of primary and recurrent recurrence. Nippon Igaku Hoshasen Gakkai Zasshi 58:
bronchogenic malignancies. Cancer 75:2345–2350 18–24
Hatlevoll R, Karlsen KO, Skovlund E (1999) Endobronchial Kopelson G, Choi NCH (1980) Radiation therapy for post-
radiotherapy for malignant bronchial obstruction or recur- operative local-regionally recurrent lung cancer. Int J Radiat
rence. Acta Oncol 38:999–1004 Oncol Biol Phys 6:1503–1506
Hauswald H, Stoiber E, Rochet N, Lindel K, Grehn C, Becker Kramer GWPM, Gans S, Ullmann E, van Meerbeck JP,
HD, Debus J, Harms W (2010) Treatment of recurrent Legrand C, Leer JWH (2004) Hypofractionated external
bronchial carcinoma: the role of high-dose-rate endoluminal beam radiotherapy as retreatment for symptomatic non-
brachytherapy. Int J Radiat Oncol Biol Phys 77:373–377 small-cell lung carcinoma: an effective treatment? Int J
Hayakawa K, Mitsuhashi N, Saito Y, Nakayama Y, Furuta M, Radiat Oncol Biol Phys 58:1388–1393
Sakurai H, Kawashima M, Ohno T, Nasu S, Niibe H (1999) Law MR, Henk JM, Lennox SC, Hodson ME (1982) Value of
Limited field irradiation for medically inoperable patients radiotherapy for tumour on the bronchial stump after
with peripheral stage I non-small cell lung cancer. Lung resection of bronchial carcinoma. Thorax 37:496–499
Cancer 26:137–142 Lesser T, Brenner A, Bartel M (1997) Das rezidiv beim kurativ
Hung J-J, Hsu W-H, Hsieh C-C, Huang B-S, Huang M-H, Liu operierten nicht-kleinzelligen bronchialkarzinom. Zentralbl
J-S, Wu Y-C (2009) Post-recurrence survival in completely Chir 122:642–648
resected stage I non-small cell lung cancer with local Leung J, Ball D, Worotniuk T, Laidlaw C (1995) Survival
recurrence. Thorax 64:192–196 following radiotherapy for post-surgical locoregional recur-
Ihde DC, Bilek FS, Cohen MH (1979) Response to thoracic rence of non-small cell lung cancer. Lung Cancer 13:
radiotherapy in patients with small cell carcinoma of the 121–127
lung after failure of combination chemotherapy. Radiology Martini N, Melamed MR (1975) Multiple primary lung cancers.
132:443–446 J Thorac Cardiovasc Surg 70:606–612
Radiation Therapy for Recurrent Disease 559

Mehta MP, Shahabi S, Jarjour NN, Kinsella TJ (1989) experience in eighty patients. J Thorac Cardiovasc Surg
Endobronchial irradiation for malignant airway obstruction. 117:1095–1101
Int J Radiat Oncol Biol Phys 17:847–851 Salazar OM, Yee GJ, Slawson RG (1991) Radiation therapy for
Mendiondo OA, Dillon M, Beach LJ (1983) Endobronchial chest recurrence following induction chemotherapy inj
brachytherapy in the treatment of recurrent bronchogenic small cell lung cancer. Int J Radiat Oncol Biol Phys
carcinoma. Int J Radiat Oncol Biol Phys 9:579–582 21:645–650
Micke O, Prott FJ, Scaaher U (1995) Endoluminal HDR Seagren SL, Harrell JH, Horn RA (1985) High dose rate
brachytherapy in the palliative treatment of patients with the intraluminal irradiation in recurrent endobronchial carci-
recurrence of a non-small-cell bronchial carcinoma after noma. Chest 88:810–814
prior radiotherapy. Strahlenther Onkol 171:554–559 Shaw EG, Brindle JS, Creagan ET, Foote RL, Trastek VF,
Montebello JF, Aron BS, Manatunga AK, Horvath JL, Peyton Buskirk SJ (1992) Locally recurrentnon-small-cell lung
FW (1993) The reirradiation of recurrent bronchogenic cancer after complete surgical resection. Mayo Clin Proc
carcinoma with external beam irradiation. Am J Clin Oncol 67:1129–1133
16:482–488 Sibley GS, Jamieson TA, Marks LB, Anscher MS, Prosnitz LR
Morita K, Fuwa N, Suzuki Y, Nishio M, Sakai K, Tamaki Y, (1998) Radiotherapy alone for medically inoperable stage I
Niibe H, Chujo M, Wada S, Sugawara T, Kita M (1997) non-small-cell lung cancer: The Duke experience. Int J
Radical radiotherapy for medically inoperable non-small Radiat Oncol Biol Phys 40:149–154
cell lung cancer in clinical stage I: retrospective analysis of Sutedja G, Baris G, Schaake-Koning C, van Zandwijk N (1992)
149 patients. Radiother Oncol 42:31–36 High dose rate brachytherapy in patients with local
Mountain CF (1986) A new international staging system for recurrences after radiotherapy of non-small cell lung cancer.
lung cancer. Chest 89:225S–233S Int J Radiat Oncol Biol Phys 24:551–553
Naruke T, Goya T, Tsuchiya R, Suemasu K (1988) Prognosis Suzuki T, Horio H, Sakaguchi K, Yamamoto M (2007)
and survival in resected lung carcinoma based on the new Lobectomy for local recurrence of lung cancer after
international staging system. J Thorac Cardiovasc Surg 3-dimensional-conformal radiotherapy. Kyobu Geka 60:
96:440–447 830–833
Neri S, Kitamura J, Komatsu T, Takahashi Y, Tajeshima Y, Tada T, Fukuda H, Matsui K, Hirashima T, Hosono M, Takada
Kaji R, Hayashi M, Nishimura T, Tomii K, Katakami N, Y, Inoue Y (2005) Non-small-cell lung cancer: reirradiation
Ishihara K, Kokubo M (2009) Lobectomy for local recur- for loco-regional relapse previously treated with radiation
rence following stereotactic radiotherapy to non-small cell therapy. Int J Clin Oncol 10:247–250
lung cancer. Kyobu Geka 62:812–815 Voltolini L, Paladini P, Luzzi L, Ghiribelli C, Di Bisceglie M,
Ochs JJ, Tester WJ, Cohen MH, Lichter AS, Ihde DC (1983) Gotti G (2000) Iterative surgical resections for local
Salvage radiation therapy for intrathoracic small cell recurrent and second primary bronchogenic carcinoma.
carcinoma of the lung progressing on combination chemo- Eur J Cardiothorac Surg 18:529–534
therapy. Cancer Treat Rep 67:1123–1126 Watanabe Y, Shimizu J, Oda M, Tatsuzawa Y, Hayashi Y, Iwa
Okamoto Y, Murakami M, Yoden E, Sasaki R, Okuno Y, T (1992) Second surgical intervention for recurrent and
Nakajima T, Kuroda Y (2002) Reirradiation for locally second primary bronchogenic cracinoma. Scand J Thorac
recurrent lung cancer previously treated with radiation Cardiovasc Surg 26:73–78
therapy. Int J Radiat Oncol Biol Phys 52:390–396 Westeel V, Choma D, Clement F, Woronoff-Lemsi M-C, Pugin
Ono R, Egawa S, Suemasu K, Sakura M, Kitagawa T (1991) J-F, Dubiez A, Depierre A (2000) Relevance of an intensive
Radiotherapy in inoperable stage I lung cancer. Jpn J Clin postoperative follow-up after surgery for non-small cell
Oncol 21:125–128 lung cancer. Ann Thorac Surg 70:1185–1190
Ornadel D, Duchesne G, Wall P, NgA HetzelM (1997) Defining Wu K-L, Jiang G-L, Qian H, Wang L-J, Yang H-J, Fu X-L,
the roles of high dose rate endobronchial brachytherapy and Zhao S (2003) Three-dimensional conformal radiotherapy
laser resection for recurrent bronchial malignancy. Lung for locoregionally recurrent lung carcinoma after external
Cancer 16:203–213 beam irradiation: a prospective phase I-II clinical trial. Int J
Pairolero PC, Williams DE, Bergstralh EJ, Piehler JM, Bernatz Radiat Oncol Biol Phys 57:1345–1350
PE, Payne WS (1984) Postsurgical stage I bronchogenic International Agency for Research on Cancer (IARC) (2010).
carcinoma: morbid implications of recurrent disease. Ann http://globocan.iarc.fr
Thorac Surg 38:331–336 Yano T, Hara N, Ichinose Y, Asoh H, Yokoyama H, Ohta M,
Poltinnikov IM, Fallon K, Xiao Y, Reiff JE, Curran WJ Jr, Werner- Hata K (1994) Local recurrence after complete resection for
Wasik M (2005) Combination of longitudinal and circimfer- nonsmall-cell carcinoma of the lung. Significance of local
ential three-dimensional esophageal dose distribution predicts control by radiation treatment. J Thorac Cardiovasc Surg
acute esophagitis in hypofractionated reirradiation of patients 10:8–12
with non-small-cell lung cancer treated in stereotactic body Zorlu AF, Selek U, Emri S, Gurkayanak M, Akyol FH (2008)
frame. Int J Radiat Oncol Biol Phys 62:652–658 Second line palliative endobronchail radiotherapy with
Regnard JF, Icard P, Magdeleinat P, Jauffret B, Farés E, HDR Ir 192 in recurrent lung carcinoma. Yonsei Med J
Levasseur P (1999) Completion pneumonectomy: 49:620–624
Radiation Therapy for Metastatic Disease
Dirk Rades

Contents Abstract
Bone metastases occur in up to 40% of NSCLC
1 Bone Metastases ....................................................... 562 patients. If associated with pathological fractures or
1.1 Background ................................................................ 562 metastatic spinal cord compression (MSCC), they are
1.2 Radiation Therapy of Bone Metastases.................... 562
1.3 Radiation Therapy of Bone Metastases: Meta-
considered ‘‘complicated’’ lesions. Otherwise, they
Analyses..................................................................... 563 are considered ‘‘uncomplicated’’. 1x8 Gy of radiation
1.4 Re-irradiation (Re-RT) for Recurrent Bone Pain..... 564 therapy (RT) can be considered the standard treat-
1.5 Complicated Bone Metastases .................................. 564 ment of most uncomplicated painful bone metastases.
2 Metastatic Spinal Cord Compression ................... 564 Single-fraction RT requires re-RT more often than
2.1 Background ................................................................ 564 multi-fraction RT regimens. However, re-RT after
2.2 Radiation Therapy of Metastatic Spinal Cord
1x8 Gy is safe and effective. If re-RT is required after
Compression .............................................................. 565
2.3 Radiation Therapy Preceded by Decompressive longer-course RT with 10x3 Gy or 20x2 Gy, the
Surgery ....................................................................... 565 second RT course should be delivered using high-
2.4 Re-irradiation for a Local Recurrence of Metastatic precision techniques. For a pathological fracture,
Spinal Cord Compression.......................................... 566
surgical stabilisation followed by RT should be
3 Brain Metastases...................................................... 566 performed. Remineralization of the osteolytic bone is
3.1 Background ................................................................ 566 better after multi-fraction RT. For MSCC, short-
3.2 Radiation Therapy of Multiple Brain Metastases .... 566
3.3 Radiation Therapy of 1–3 Brain Metastases ............ 568 course RT is as effective as longer RT programs
regarding motor function. Local control of MSCC is
4 Liver and Lung Metastases .................................... 568
better after longer-course RT. Patients with MSCC
4.1 Background ................................................................ 568
4.2 Stereotactic Body Radiation Therapy of Liver from NSCLC and a favorable survival prognosis
Metastases .................................................................. 569 may be considered candidates for decompressive
4.3 Stereotactic Body Radiation Therapy of Lung surgery followed by longer-course RT. Decompres-
Metastases .................................................................. 570
sive surgery would also be the first choice for a
References.......................................................................... 570 local recurrence of MSCC after longercourse RT.
A recurrence after short-course RT can be safely
treated with another short-course of RT. Brain
metastases occur in 20–40% of cancer patients.
NSCLC is the most common primary tumor and
which accounts for at least 40% or more of these
patients. Most patients have multiple (4) lesions and
usually receive whole-brain radiotherapy (WBRT)
D. Rades (&)
Department of Radiation Oncology, alone. Patients with a poor survival prognosis should
University of Lübeck, Lübeck, Germany receive 5x4 Gy in one week, whereas patients with a
e-mail: Rades.Dirk@gmx.net

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 561
DOI: 10.1007/174_2011_275, Ó Springer-Verlag Berlin Heidelberg 2011
562 D. Rades

more favourable prognosis are candidates for longer- metastases plus a margin of 2–3 cm. Overall response
course WBRT. Patients with 1–3 brain metastases rates of 50–90% were reported, and complete
have a considerably better survival prognosis response rates ranged from 10 to 50% (Amouzegar-
and may benefit from more intensive treatments Hashemi et al. 2008; Bone Pain Trial Working Party
including radiosurgery or surgery. WBRT in addition 1999; Foro Arnalot et al. 2008; Gaze et al. 1997;
to radiosurgery or surgery leads to improved local Hartsell et al. 2005; Hoskin et al. 1992; Jeremic et al.
control. The non-invasive regimen radiosurgery + 1998; Koswig and Budach 1999; Nielsen et al. 1998;
WBRT is at least as effective as surgery + WBRT, Niewald et al. 1996; Okawa et al. 1988; Rasmusson
and, therefore, preferable. Most data of SBRT of liver et al. 1995; Roos et al. 2005; Steenland et al. 1999;
metastases have been obtained from colorectal cancer Tong et al. 1982). During RT, the phenomenon of
patients bur can be ‘‘extrapolated’’ to liver metastases pain flare, a transient increase of bone pain, may
from NSCLC. SBRT if used for a limited number of occur. The pain flare rate reported to be 14–44%
liver metastases can lead to high local control rates can be reduced by prophylactic administration of
and considered a reasonable alternative to other local dexamethasone (Chow et al. 2007a). Several ran-
treatments. If SBRT is used for the treatment of lung domized trials compared different fractionation
metastases, lung tissue density correction during regimens with respect to response, need for re-RT
treatment planning is very important. In the treatment because of recurrent pain, and pathological fractures
of a limited number of lung metastases, SBRT can following RT. Two randomized trials compared
result in excellent local control rates and median different single-fraction regimens. In 1992, Hoskin
survival times of more than three years. In summery, et al. showed in 270 patients that 1 9 8 Gy was
radiation therapy if appropriately tailored to the superior to 1 9 4 Gy for overall pain response
individual patient with metastasis from NSCLC can (69 vs. 44%, P \ 0.001) and need for re-RT (9 vs.
provide valuable results in terms of relief of symp- 20%, P \ 0.05) (Hoskin et al. 1992). Complete pain
toms and improvement of the patient’s prognosis. relief was similar in both groups (39 vs. 36%,
P [ 0.05). In 1998, Jeremic et al. compared 1 9 4,
1 9 6 and 1 9 8 Gy in 327 patients (Jeremic et al.
1998). The rates of overall response were 59% after
1 Bone Metastases 1 9 4 Gy, 73% after 1 9 6 Gy and 78% after
1 9 8 Gy. Complete pain relief was achieved in 21,
1.1 Background 27 and 32% of patients, respectively. Results after
1 9 6 and 1 9 8 Gy were not significantly dif-
Bone metastases occur in up to 40% of lung cancer ferent. 8 Gy may be considered as probably ‘‘low-
patients during the course of their disease (Coleman est’’ optimal single-fraction RT.
2006). If bone metastases lead to pathological frac- Also, several randomized trials compared single-
tures or metastatic spinal cord compression, they are fraction RT of 1 9 8 Gy to a multi-fraction short-
considered ‘‘complicated’’ lesions. Otherwise, they course regimen such as 5 9 4 or 6 9 4 Gy given in
are considered ‘‘uncomplicated’’. The survival prog- five or six working days. Two large trials are men-
nosis depends on the type of primary tumor. Patients tioned in detail. A randomized trial from the United
with bone metastases from lung cancer have an Kingdom compared 1 9 8 to 5 9 4 Gy in a series of
unfavorable prognosis of a few months (Coleman 761 patients (Bone Pain Trial Working Party 1999).
2006). However, there are long-term survivors as well No significant difference was observed between 1 9 8
(Coleman and Rubens 1987). and 5 9 4 Gy with respect to overall pain response
(72 vs. 68%, P [ 0.05), complete pain relief (52 vs.
1.2 Radiation Therapy of Bone 51%, P [ 0.05) and post-RT pathological fractures
Metastases (2 vs. \1%, P = 0.2). Re-irradiation for another
episode of pain in the same area was required more
Radiation therapy (RT) alone is the standard treat- often after 1 9 8 than 5 9 4 Gy (23 vs. 10%,
ment for uncomplicated painful bone metastases. P \ 0.001). A randomized trial of 1,171 patients from
The treatment volume generally encompasses the The Netherlands compared 1 9 8 to 6 9 4 Gy
Radiation Therapy for Metastatic Disease 563

Fig. 1 Example of stereotactic body radiation therapy (SBRT) of a bone metastasis in the vertebral column. SBRT provides
excellent sparing of spinal cord and kidney

(Steenland et al. 1999). No significant difference was 1.3 Radiation Therapy of Bone
observed between 1 9 8 and 6 9 4 Gy with respect to Metastases: Meta-Analyses
overall pain response (72 vs. 69%, P = 0.24) and
complete pain relief (37 vs. 33%, P [ 0.05). The rate In 2003, Wu et al. presented a meta-analysis of eight
of post-RT pathological fractures was significantly randomized trials with 3,260 patients that compared
higher in the 1 9 8 Gy group although being consid- 1 9 8 Gy to multi-fraction regimens ranging from
erably low (4 vs. 2%, P \ 0.05). Re-irradiation was 5 9 4 to 10 9 3 Gy (Wu et al. 2003). In the treatment
required more often after 1 9 8 Gy (25 vs. 7%, per-protocol analysis (N = 2,683), 39% of patients after
P \ 0.001). Several other randomized trials compared 1 9 8 Gy and 50% of patients after multi-fraction RT,
single-fraction RT of 1 9 8 Gy to multi-fraction respectively, achieved complete pain relief (relative risk
longer-course RT of 10 9 3 Gy in two weeks. In the [RR] 0.98; 95%-confidence interval [CI] 0.89-1.07;
largest trial (N = 888), no significant difference was P = 0.6). The overall response rates were 73 and 73%,
observed between 1 9 8 and 10 9 3 Gy with respect respectively (RR 1.00; 95%-CI 0.95-1.04; P = 0.9).
to overall pain response (65 vs. 66%, P = 0.6), com- Wu et al. did not report the need for re-irradiation and
plete pain relief (15 vs. 18%, P [ 0.05) and post-RT post-RT pathological fracture rates. Similar results
pathological fractures (5 vs. 4%, P [ 0.05) (Hartsell were reported by Sze et al. for 3,621 patients from 12
et al. 2005). The need for re-irradiation for another randomized trials (Sze et al. 2003). The rates of complete
episode of pain in the same area was greater after 1 9 8 pain relief were 34% after single-fraction RT and 32%
than 10 9 3 Gy (18 vs. 9%, P \ 0.001). Randomized after multi-fraction RT (odds ratio [OR] 1.10; 95%-CI
trials that compared different multi-fraction regimens 0.94-1.30, P [ 0.05). The overall pain response rates
such as 5 9 4, 5 9 5, 10 9 3, or 15 9 2 Gy showed were 60 and 59%, respectively (OR 1.03; 95%-CI
that no particular regimen was superior to another 0.90-1.19; P [ 0.05). The re-irradiation rates were
regimen (Niewald et al. 1996; Okawa et al. 1988; 22% after single-fraction and 7% after multi-fraction RT
Rasmusson et al. 1995; Tong et al. 1982). (OR 3.44; 95%-CI 2.67-4.43; P \ 0.05). The post-RT
564 D. Rades

pathological fracture rates were 3.0 and 1.6%, respec- the tumour tissue, post-operative RT is required to
tively (OR 1.82; 95%-CI 1.06-3.11; P \ 0.05). This avoid a recurrence and slackening or dislocation of
may be explained by the fact that 10 9 3 Gy can lead to the osteosynthetic material. Another important goal of
a significantly higher increase in bone density than radiation therapy is the remineralization of the oste-
1 9 8 Gy (Koswig and Budach 1999). In 2007, Chow olytic bone, which appears better after multi-fraction
et al. presented a meta-analysis of 5,000 patients from 16 than after single-fraction radiation therapy (Koswig
randomized trials (Chow et al. 2007b). The overall and Budach 1999). Because significant remineraliza-
pain response rates were 58% after single-fraction RT tion can only be expected several months after RT,
(mostly 1 9 8 Gy) and 59% after multi-fraction RT patients with a favorable survival prognosis may not
(mostly 5 9 4 or 10 9 3 Gy) (OR 0.99; 95%-CI be treated with single-course RT. However, many
0.95-1.03; P = 0.60). Complete pain relief was patients with bone metastases from non-small cell
achieved in 23 and 24% (558/2,351) of patients, lung cancer have a poor survival prognosis and,
respectively (OR 0.97; 95%-CI 0.88-1.06; P = 0.51). therefore, are candidates for single-fraction RT. The
The re-irradiation rates were 20% after single-fraction survival prognosis of patients with bone metastases
and 8% (158/2,032) after multi-fraction RT, respectively can be estimated with scoring systems (Katagiri et al.
(OR 2.50; 95%-CI 1.76-3.56; P \ 0.001). The rates of 2005; Van der Linden et al. 2005). Another possible
pathological fractures following RT were not signifi- complication is metastatic spinal cord compression,
cantly different, 3.2 and 2.8%, respectively (OR 1.10; which is covered below.
95%-CI 0.61-1.99; P = 0.75). This result is in contrast
to the data of the meta-analysis of Sze et al. (2003). Thus,
it remains unclear whether single-fraction RT is really 2 Metastatic Spinal Cord
associated with a higher rate of pathological fractures. Compression

2.1 Background
1.4 Re-irradiation (Re-RT) for Recurrent
Bone Pain Metastatic Spinal Cord Compression (MSCC) occurs
in 5–10% of all patients with cancer during the course
Re-RT after single-fraction RT is safe and effective of their disease (Loblaw et al. 2003; Bach et al. 1990).
(Jeremic et al. 1999; Mithal et al. 1994). Acute toxicity Lung cancer is one of the most common primary
of re-RT has been reported to not exceed grade 2. In a tumors in patients with MSCC accounting for about
retrospective study of 105 patients, the overall pain 20% of these patients. Despite improved treatment
response rate after re-RT was 87% (Jeremic et al. 1999). approaches, the treatment of MSCC still is a chal-
Eight patients received a third course of RT to the same lenge, in particular when one intends to tailor the
region. Response was observed in seven patients. treatment to each patient. The most common symp-
In another study of 109 patients, the overall response tom of MSCC is back pain (70–96%) followed by
rate to re-RT was 74% (Mithal et al. 1994). If re-RT motor deficits (61–91%), sensory deficits (46–90%),
is required after longer-course RT with 10 9 3 or and dysfunction of bladder or bowel control
20 9 2 Gy, the second course of RT should be delivered (40–57%) (Bach et al. 1990; Gilbert et al. 1978).
using high-precision techniques (stereotactic body RT, If pain is the only symptom without neurologic defi-
radiosurgery, intensity-modulated RT, proton beams) to cits, the situation can be described as impending
better spare healthy tissues and reduce the risk of late MSCC. Dysfunction of bladder or bowel control
toxicity. An example of stereotactic body RT from the occurs late and requires surgical intervention when-
University of Lubeck, Germany, is given in Fig. 1. ever possible. However, radiation RT alone is the
most common treatment for MSCC. It should be
1.5 Complicated Bone Metastases supplemented by administration of dexamethasone
whenever possible. To avoid severe side affects such
In case of a pathological fracture, surgical stabilisa- as gastrointestinal bleeding and psychosis, an inter-
tion should be performed whenever possible. Because mediate dose of 12–40 mg/day with a taper during or
surgery usually does not result in complete removal of immediately after RT is recommended.
Radiation Therapy for Metastatic Disease 565

2.2 Radiation Therapy of Metastatic tumors. Besides functional outcome, local control
Spinal Cord Compression of MSCC is another important endpoint, in partic-
ular for long-term survivors. These patients may
The treatment volumes of RT for MSCC usually live long enough to experience such a recurrence.
encompass one to two normal vertebral bodies above In a prospective study, longer-course RT resulted in
and below the metastatic lesions. If only a limited significantly better 1-year local control than short-
number of vertebrae are involved and the survival course RT (81 vs. 61%, P = 0.005) (Rades et al.
prognosis is favorable, RT can be administered to the 2011b). Thus, short-course RT appears preferable
involved vertebrae plus a safety margin of 1–2 cm for patients with a poor expected survival including
based on computed tomography-based treatment most NSCLC patients. Patients with a better
planning. The most appropriate RT regimen depends survival prognosis may benefit from longer-course
on the patient’s survival prognosis and the endpoints. RT in terms of better remineralization of the oste-
Improvement of motor function is generally consid- olytic bone and better local control of MSCC. The
ered the most relevant endpoint. Four published pro- survival prognosis of MSCC patients can be esti-
spective studies compared different RT regimens with mated with a validated scoring system (Rades et al.
respect to functional outcome (Rades et al. 2004, 2008a; 2010a). Patients with a very favorable
2011; Maranzano et al. 2005, 2009). In 2004, a non- prognosis may be candidates for high-precision RT,
randomized prospective study of 214 patients from which allows better sparing of the surrounding
Germany compared 10 9 3 Gy in two weeks and normal tissues and/or a dose escalation to the
20 9 2 Gy in four weeks (Rades et al. 2004). The metastatic site.
rates of improvement of motor function (43 vs. 41%,
P = 0.80) and post-RT ambulatory status (60 vs.
64%, P = 0.71) were similar in both groups. In 2005, 2.3 Radiation Therapy Preceded
a randomized trial of 276 patients from Italy by Decompressive Surgery
compared 2 9 8 Gy and a split-course regimen
(3 9 5 Gy in three days followed by four-day-rest The major advantages of spinal surgery when com-
and 5 9 3 Gy in one week) and found both schedules pared with RT are immediate decompression of the
similarly effective (Maranzano et al. 2005). In 2009, spinal cord and direct mechanical stabilization of the
another randomized study from Italy demonstrated spine. Indications for spinal surgery include intras-
that 1 9 8 and 2 9 8 Gy in eight days resulted in pinal bony fragments, spinal instability, impending or
similar functional outcome (Maranzano et al. 2009). present sphincter dysfunction, no response to previous
Recently, a prospective study of 265 patients from radiotherapy treatment, and a recurrence of MSCC
Germany and The Netherlands comparing short- after longer-course RT. The benefit of surgery plus
course (1 9 8 or 5 9 4 Gy) to longer-course RT RT compared to RT alone is still controversial.
(10 9 3, 15 9 2.5 or 20 9 2 Gy) suggested both A small randomized study of 101 patients found a
regimens to be similarly effective with respect to benefit for surgery followed by 10 9 3 Gy compared
functional outcome (Rades et al. 2011b). Motor to 10 9 3 Gy alone (Patchell et al. 2005). Signifi-
function improved in 37% after short-course and 39% cantly more patients in the surgery group were able to
after longer-course RT (P = 0.95). walk after treatment (84 vs. 57%, P = 0.001).
One retrospective study that focused particularly Patients who received surgery maintained the ability
on patients with MSCC from NSCLC was reported to walk for a longer period (122 vs. 13 days,
in 2006 (Rades et al. 2006). That study compared P = 0.003). However, several researchers suggested
short-course and longer-course RT with respect to that the results may have been confounded due to
functional outcome. Improvement of motor function methodological problems (Kunkler 2006; Knisely and
was observed in 15% of patients after short-course Strugar 2006). Furthermore, the study appeared sta-
and 13% after longer-course RT (P = 0.87). tistically underpowered, and only very selected
Furthermore, these results showed that patients with patients with an expected survival of C3 months,
MSCC from NSCLC have a worse functional out- a good performance status, and involvement of
come after RT than patients with other primary only one spinal area were included. In a recent
566 D. Rades

multi-national matched-pair analysis of 324 MSCC precision RT (Milker-Zabel et al. 2003; Gerszten
patients, improvement of motor function occurred in et al. 2007).
27% after surgery plus RT and 26% after RT alone
(P = 0.92) (Rades et al. 2010b). Post-treatment
ambulatory rates were 69 and 68%, respectively 3 Brain Metastases
(P = 0.99). An additional matched-pair analysis
performed by the same group for tumors considered 3.1 Background
unfavorable with respect to response to RT alone
(including NSCLC) suggested that these patients do Brain metastases occur in 20–40% of all cancer
benefit from decompressive surgery in addition to RT patients during the course of their disease (Khuntia
(Rades et al. 2011c). Motor function improved in 28 et al. 2006). NSCLC is the most common primary
and 19% of patients, respectively (P = 0.024). tumor, which accounts for 40% or more of all patients
NSCLC patients with a relatively favorable survival with brain metastases. The number of brain metastases
prognosis may be considered candidates or decom- is an important prognostic factor. About 60% of
pressive surgery followed by longer-course RT. patients have multiple (C4) lesions, and about 40%
have a limited number of 1–3 lesions or a single lesion.
A solitary lesion is defined as a single lesion without
2.4 Re-irradiation for a Local Recurrence additional extracerebral metastases. Headache is
of Metastatic Spinal Cord the most common clinical symptom. Depending on
Compression the metastatic site, symptoms may vary including
seizures, vision disturbances, hearing problems, nau-
A local recurrence of MSCC in the previously irra- sea, motor deficits, and others.
diated area of the spinal cord may occur. 1-year
local failure rates of 19% after longer-course and
39% after short-course RT have been reported 3.2 Radiation Therapy of Multiple Brain
(Rades et al. 2011b). Decompressive surgery would Metastases
be the first choice in patients who develop a recur-
rence of MSCC in the previously irradiated region. Patients with multiple brain metastases usually receive
However, the indication for surgery is limited to whole-brain radiotherapy (WBRT) plus dexametha-
10–15% of these patients and re-irradiation may be sone (12–40 mg/day with a taper during or immedi-
the only alternative option (Patchell et al. 2005; ately after WBRT) (Khuntia et al. 2006). Most patients
Prasad and Schiff 2005). However, a second series with brain multiple metastases have a poor survival
may result in a relatively high cumulative biologi- prognosis. This accounts particularly for patients with
cally effective dose (BED) and an increased risk of brain metastases from NSCLC. The median survival
radiation myelopathy. Re-irradiation seems well of untreated patients with multiple lesions is about one
tolerated if the cumulative BED is B120 Gy2, which month (Zimm et al. 1981). Even with WBRT, these
is the case if two series of short-course RT are patients have a median life expectancy of only a few
administered (Rades et al. 2008b). Re-irradiation months (Sundstrom et al. 1998). Thus, short-course
with 1 9 8, 5 9 3 or 5 9 4 Gy proved to be effec- WBRT such as 5 9 4 Gy in 1 week would be pref-
tive (Rades et al. 2008b, 2005). Improvement of erable, if it provided similar outcomes as longer
motor function occurred in 40% of the re-irradiated WBRT programs such as 10 9 3 Gy in 2 weeks and
patients, and progression of motor deficits was 20 9 2 Gy in 4 weeks. A few studies compared short-
stopped in another 45% (Rades et al. 2005). After course to longer-course WBRT for multiple brain
longer-course RT with a higher BED (75 Gy2 for metastases (Harwood and Simson 1977; Priestman
10 9 3 Gy and 80 Gy2 for 20 9 2 Gy), the cumu- et al. 1996; Borgelt et al. 1980; Borgelt et al. 1981;
lative BED is likely to exceed 120 Gy2 (Rades et al. Chatani et al. 1994). Whereas most studies found no
2008b). In such a situation, high-precision RT significant differences in survival, one prospective
should be used. Improvement of neurological deficits study suggested a marginally better median survival
was reported in 42 and 84% of patients after high- after long-course WBRT with 10 9 3 Gy than after
Radiation Therapy for Metastatic Disease 567

Fig. 2 Example of linear accelerator based stereotactic radiosurgery of a single brain metastasis

short-course WBRT with 2 9 6 Gy (2.8 vs. WBRT with lower doses per fraction, as it has been
2.5 months, P = 0.04) (Priestman et al. 1996). Only suggested that the risk of radiation induced neuro-
one study focused on NSCLC patients and compared cognitive deficits is lower with doses per fraction of
5 9 4 Gy (n = 140) to 10 9 3 or 20 9 2 Gy \3Gy (De Angelis et al. 1989). The survival prog-
(n = 262) with respect to survival (Rades et al. nosis of patients with brain metastases can be
2007a). Of the 404 patients, 16% had a single lesion. estimated with the help of prognostic scores (Gaspar
The survival rates at 6, 12, and 24 months were 40, 16, et al. 1997; Rades et al. 2011a). The attempt to
and 6%, respectively, after 5 9 4 Gy, and 30, 17, and improve the survival after WBRT with the addition of
10%, respectively, after longer-course WBRT chemotherapy has failed (Antonadou et al. 2002;
(P = 0.55). Thus, most patients with multiple brain Postmus et al. 2000; Mornex et al. 2003). However,
metastases from NSCLC appear not to benefit from data from a very limited number of patients suggested
longer-course WBRT. Patients with a favorable sur- median survival times of more than a year with the use
vival prognosis may be candidates for longer-course of tyrosine-kinase inhibitors (Ma et al. 2009).
568 D. Rades

3.3 Radiation Therapy of 1–3 Brain (Aoyama et al. 2006); 1-year local control rates were
Metastases 53 and 24%, respectively (P \ 0.001). A more recent
randomized trial of 359 patients compared radiosur-
Patients with 1–3 brain metastases have a consider- gery or surgery alone to radiosurgery or surgery plus
ably better survival prognosis than patients with C4 WBRT (Kocher et al. 2011). Survival was similar in
lesions and may benefit from a more intensive treat- both groups (P = 0.89). The addition of WBRT
ment including radiosurgery or surgery. An example improved local control after surgery (P \ 0.001) and
of stereotactic radiosurgery from the University of radiosurgery (P = 0.40). Local control is an impor-
Lubeck, Germany, is given in Fig. 2. tant endpoint, because an intracerebral recurrence
For patients with a single lesion, three small (and not the addition of WBRT) is considered the
randomized trials compared WBRT alone to most relevant factor for a post-treatment decline in
WBRT ? surgery (Patchell et al. 1990; Vecht et al. neurocognitive function (Meyers et al. 2004; Aoyama
1993; Mintz et al. 1996). Patchell et al. reported a et al. 2007). Aoyama et al. (2007) showed in a sub-
longer median survival time (9.2 vs. 3.5 months, group analysis of their randomized trial that the
P \ 0.01) and fewer local recurrences (20 vs. 52%, neurocognitive function at one and two years was
P \ 0.02) after combined treatment than WBRT significantly better in those patients receiving WBRT
alone in 48 patients (Patchell et al. 1990). Median in addition to radiosurgery.
survival times in the study of Vecht et al. (N = 63) In two retrospective studies that compared radio-
were 10 months after WBRT ? surgery and surgery ? WBRT to surgery ? WBRT, all patients
6 months after WBRT alone (P = 0.04) (Vecht et al. had received WBRT (Schöggl et al. 2000; Rades
1993). Mintz et al. (N = 84) did not find a significant et al. 2009). Schöggl et al. (2000) who compared
difference in median survival (5.6 vs. 6.3 months, both treatment regimens in 133 patients with a single
P = 0.24) (Mintz et al. 1996). In a more recent ret- brain metastasis, reported median survival times of
rospective study of 195 patients, WBRT ? surgery 12 months in the radiosurgery and 9 months in the
resulted in better 1-year survival (48 vs. 26%, surgery group (P = 0.19). Local control was sig-
P \ 0.001) and 1-year intracerebral control (57 vs. nificantly (P \ 0.05) better in the radiosurgery
24%, P \ 0.001) than WBRT alone (Rades et al. group. Rades et al. (2009) reported a matched-pair
2008c). Considering the available data, WBRT ? analysis of 104 patients with 1–3 brain metastases.
surgery is superior to WBRT alone. In a randomized The 1-year local control rates were 87% after radi-
trial of 95 patients that compared surgery alone to osurgery ? WBRT and 66% after surgery ? WBRT
surgery ? WBRT, surgery ? WBRT resulted in (P = 0.021). 1-year survival rates were 56 and 41%,
better 1-year intracerebral control (82 vs. 30%, respectively (P = 0.08). The non-invasive regimen
P \ 0.001), whereas median survival was similar in radiosurgery ? WBRT appears at least as effective
both groups (Patchell et al. 1998). as surgery ? WBRT, and, therefore, appears pref-
In patients with 1–3 lesions, radiosurgery alone erable to surgery ? WBRT for both patients with a
was reported to be superior to WBRT alone single lesion and patients with 1–3 lesions. WBRT
with respect to 1-year local control (64 vs. 26%, should be performed as longer-course WBRT, pref-
P \ 0.001) and 1-year survival (52 vs. 33%, erably with doses per fraction of \3 Gy, in order to
P = 0.045) in a retrospective series of 186 patients further reduce the risk of post-treatment neurocog-
(Rades et al. 2007b). Another retrospective study of nitive deficits.
144 patients compared radiosurgery alone to WBRT
plus radiosurgery (Rades et al. 2008d). 1-year survival
rates were 53% after radiosurgery alone and 56% 4 Liver and Lung Metastases
after WBRT ? radiosurgery (P = 0.24); 1-year local
control rates were 66 and 87% (P = 0.003), respec- 4.1 Background
tively. In a randomized trial of 132 patients with 1–4
brain metastases from Japan, the 1-year survival rates Because systemic therapies have considerably
were 39% in the WBRT ? radiosurgery group and improved in the treatment of metastatic NSCLC, in
28% in the radiosurgery alone group (P = 0.42) particular in the case of liver and lung metastases, the
Radiation Therapy for Metastatic Disease 569

Fig. 3 Example of stereotactic body radiation therapy (SBRT) of a single pulmonary lesion providing excellent sparing of heart
and lungs

role of conventional RT has become less important. more than 50% of the liver should receive 15 Gy, and
This accounts in particular for whole-liver irradiation no more than 30% should receive 21 Gy. In case of a
(Timmerman et al. 2009). However, if only a limited single-fraction treatment, the doses are 12 and 7 Gy,
number of liver or lung metastases require treatment, respectively. A few small studies are available that
stereotactic body radiation therapy (SBRT) may be an investigated local control and survival after SBRT for
option, particularly if a resection of the metastases is liver metastases (Wulf et al. 2001; Mendez Romero
not possible or refused by the patient. et al. 2006; Herfarth et al. 2001; Katz et al. 2007;
Kavanagh et al. 2006). Two studies administered three
fractions of 10 (Wulf et al. 2001) and 12.5 Gy
4.2 Stereotactic Body Radiation Therapy (Mendez Romero et al. 2006), respectively. The 2-year
of Liver Metastases local control rates were 61 and 82%, respectively, and
the 2-year survival rates were 41 and 50%, respec-
Most data regarding SBRT of liver metastases have tively. Herfarth et al. presented a dose-escalation
been obtained from colorectal cancer patients and not study of single-fraction SBRT (15–26 Gy) and
from NSCLC patients. However, ‘‘extrapolation’’ of achieved a 67% local control rate at 18 months
the data from liver metastases from colorectal cancer (Herfarth et al. 2001). Katz et al. (2007) used dif-
to metastases from NSCLC appears possible. Gener- ferent fractionation regimens (30–55 Gy given in
ally, the size of liver metastases treated with SBRT 5–15 fractions); at 20 months, local control and
should be \5 cm, although fractionated SBRT has survival rates were 57 and 37%, respectively.
been used for lesions of up to 10 cm (Timmerman Kavanagh et al. reported an actuarial local control
et al. 2009). Radiation-induced liver disease (RILD) rate of 93% after SBRT with 3 9 20 Gy for 1–3
including symptoms such as hepatomegaly, ascites liver metastases, which they considered safe (Kava-
and elevated liver transaminases is a serious toxicity nagh et al. 2006). Thus, SBRT if used for a limited
following liver irradiation and may also occur number of liver metastases can lead to high local
following SBRT (Hoyer et al. 2006; Blomgren et al. control rates and can be considered a reasonable
1995). If the treatment is given in three fractions, no alternative to other local treatments.
570 D. Rades

4.3 Stereotactic Body Radiation Therapy Aoyama H, Shirato H, Tago M et al (2006) Stereotactic
of Lung Metastases radiosurgery plus whole-brain radiation therapy vs. stereo-
tactic radiosurgery alone for treatment of brain metastases.
A randomized controlled trial. JAMA 295:2483–2491
An example of SBRT for a pulmonary lesion from the Aoyama H, Tago M, Kato N et al (2007) Neurocognitive
University of Lubeck, Germany, is given in Fig. 3. function of patients with brain metastasis who received
If SBRT is used for the treatment of lung metastases, either whole brain radiotherapy plus stereotactic radiosur-
gery or radiosurgery alone. Int J Radiat Oncol Biol Phys
lung tissue density correction during treatment plan- 68:1388–1395
ning is very important (Timmerman et al. 2009). Bach F, Larsen BH, Rohde K et al (1990) Metastatic spinal
Toxicities which may occur after RT of lung tumors cord compression. Occurrence, symptoms, clinical presen-
and metastases include pneumonitis, decline in tations, and prognosis in 398 patients with spinal cord
compression. Acta Neurochir 107:37–43
pulmonary function, hemoptysis, and atelectasis. The Blomgren H, Lax I, Naslund I et al (1995) Stereotactic high
risk of such toxicities is higher for central lesions. dose fractionation radiation therapy of extracranial tumors
Because most lung metastases are located peripher- using an accelerator. Clinical experience of the first thirty-
ally, the risk of RT- related toxicity is lower than for one patients. Acta Oncol 34:861–870
Bone Pain Trial Working Party (1999) 8 Gy single fraction
primary lung tumors. Furthermore, the irradiated lung radiotherapy for the treatment of metastatic skeletal pain
volume is smaller with SBRT than with conventional (randomised comparison with a multifraction schedule over
RT. Between 1995 and 2003, several small studies 12 months of patient follow-up). Radiother Oncol 52:
with less than 25 patients were reported using a great 111–121
Borgelt B, Gelber R, Kramer S et al (1980) The palliation of
variety of different fractionation regimens (Blomgren brain metastases: final results of the first two studies by the
et al. 1995; Wulf et al. 2001; Uematsu et al. 1998; Radiation Therapy Oncology Group. Int J Radiat Oncol Biol
Nakagawa et al. 2000; Nagata et al. 2002; Hara et al. Phys 6:1–9
2002; Lee et al. 2003). The local control rates ranged Borgelt B, Gelber R, Karson M et al (1981) Ultra-rapid high
dose irradiation schedules for the palliation of brain
between 67 and 98%. In 2006, Okunieff et al. reported metastases: final results of the first two studies by the
on 50 patients who received 10 9 5 Gy of SBRT for radiation therapy oncology group. Int J Radiat Oncol Biol
1–5 pulmonary lesions (Okunieff et al. 2006). Of the Phys 7:1633–1638
total of 125 lesions, eight progressed (local control Chatani M, Matayoshi Y, Masaki N et al (1994) Radiation
therapy for brain metastases from lung carcinoma. Prospec-
rate 94%). The median overall survival time was tive randomized trial according to the level of lactate
23.4 Gy. In 2008, Norihisa et al. presented 34 patients dehydrogenase. Strahlenther Onkol 170:155–161
who received 4–5 9 12 Gy of SBRT for 1–2 lesions Chow E, Loblaw A, Harris K et al (2007a) Dexamethasone for
(Norihisa et al. 2008). The 2-year local control rate the prophylaxis of radiation-induced pain flare after palli-
ative radiotherapy for bone metastases—a pilot study.
was 90%, the 2-year overall survival rate was 84%, Support Care Cancer 15:643–647
and the median survival time was [36 months. In a Chow E, Harris K, Fan G et al (2007b) Palliative radiotherapy
most recent study presented by Rusthoven et al. trials for bone metastases: a systematic review. J Clin Oncol
(2009) the 2-year actuarial local control rate was 25:1423–1436
Coleman RE (2006) Clinical features of metastatic bone disease
96%, and the median survival time was 19 months. and risk of skeletal morbidity. Clin Cancer Res 12
For a limited number of lung metastases, SBRT can (Suppl.):6243 s–6249 s
result in excellent local control rates and median Coleman RE, Rubens R (1987) The clinical course of bone
survival times of more than three years. metastases in breast cancer. Br J Cancer 77:336–340
De Angelis LM, Mandell LR, Thaler HAT et al (1989) The role
of postoperative radiotherapy after resection of single brain
metastasis. Neurosurgery 24:798–805
References Foro Arnalot P, Fontanais AV, Galceran JC et al (2008)
Randomized clinical trial with two palliative radiotherapy
regimens in painful bone metastases: 30 Gy in 10 fractions
Amouzegar-Hashemi F, Behrouzi H, Kazemian A et al (2008) compared with 8 Gy in single fraction. Radiother Oncol
Single versus multiple fractions of palliative radiotherapy 89:150–155
for bone metastases: a randomized clinical trial in Iranian Gaspar LE, Scott C, Rotman M et al (1997) Recursive
patients. Curr Oncol 15:36–39 partitioning analysis (RPA) of prognostic factors in three
Antonadou D, Paraskevaidis M, Sarris G et al (2002) Phase II radiation therapy oncology group (RTOG) brain metastases
randomized trial of temozolomide and concurrent radio- trials. Int J Radiat Oncol Biol Phys 37:745–751
therapy in patients with brain metastases. J Clin Oncol Gaze MN, Kelly CG, Kerr GR et al (1997) Pain relief and
20:3644–3650 quality of life following radiotherapy for bone metastases:
Radiation Therapy for Metastatic Disease 571

a randomised trial of two fractionation schedules. Radiother Lee SW, Choi EK, Park HJ et al (2003) Stereotactic body frame
Oncol 45:106–116 based fractionated radiosurgery on consecutive days for
Gerszten PC, Burton SA, Ozhasoglu C et al (2007) Radiosur- primary or metastatic tumors in the lung. Lung Cancer
gery for spinal metastases: clinical experience in 500 cases 40:309–315
from a single institution. Spine 32:193–199 Loblaw DA, Laperriere NJ, Mackillop WJA (2003) Population-
Gilbert RW, Kim JH, Posner JB (1978) Epidural spinal cord based study of malignant spinal cord compression in
compression from metastatic tumor: diagnosis and treat- Ontario. Clin Oncol 15:211–217
ment. Ann Neurol 3:40–51 Ma S, Xu Y, Deng Q et al (2009) Treatment of brain metastasis
Hara R, Itami J, Kondo T et al (2002) Stereotactic single high from non-small cell lung cancer with whole brain radio-
dose irradiation of ling tumors under respiratory gating. therapy and Gefitinib in a Chinese population. Lung Cancer
Radiother Oncol 63:159–163 65:198–203
Hartsell WF, Scott CB, Bruner DW et al (2005) Randomized Maranzano E, Bellavita R, Rossi R et al (2005) Short-course
trial of short- versus long-course radiotherapy for palliation versus split-course radiotherapy in metastatic spinal cord
of painful bone metastases. J Natl Cancer Inst 97:798–804 compression: results of a phase III, randomized, multicenter
Harwood AR, Simson WJ (1977) Radiation therapy of cerebral trial. J Clin Oncol 23:3358–3365
metastases: a randomized prospective clinical trial. Int J Maranzano E, Trippa F, Casale M et al (2009) 8 Gy single-dose
Radiat Oncol Biol Phys 2:1091–1094 radiotherapy is effective in metastatic spinal cord compres-
Herfarth KK, Debus J, Lohr F et al (2001) Stereotactic single- sion: results of a phase III randomized multicentre Italian
dose radiation therapy of liver tumors: results of a phase I/II trial. Radiother Oncol 93:174–179
trial. J Clin Oncol 19:164–170 Mendez Romero A, Wunderink W, Hussain SM et al (2006)
Hoskin PJ, Price P, Easton D et al (1992) A prospective Stereotactic body radiation therapy for primary and meta-
randomised trial of 4 Gy or 8 Gy single doses in the static liver tumors: a single institution phase I–II study. Acta
treatment of metastatic bone pain. Radiother Oncol 23: Oncol 45:831–837
74–78 Meyers CA, Smith JA, Bezjak A et al (2004) Neurocognitive
Hoyer M, Henrik R, Traberg-Hansen A (2006) Phase II study function and progression in patients with brain metastases
on stereotactic body radiotherapy of colorectal metastases. treated with whole-brain radiation and motexafin gadolin-
Acta Oncol 45:823–830 ium: results of a randomized phase III trial. J Clin Oncol
Jeremic B, Shibamoto Y, Acimovic L et al (1998) 22:157–165
A randomized trial of three single-dose radiation therapy Milker-Zabel S, Zabel A, Thilmann C et al (2003) Clinical
regimens in the treatment of metastatic bone pain. Int J results of retreatment of vertebral bone metastases by
Radiat Oncol Biol Phys 42:161–167 stereotactic conformal radiotherapy and intensity-modulated
Jeremic B, Shibamoto Y, Igrutinovic I (1999) Single 4 Gy re- radiotherapy. Int J Radiat Oncol Biol Phys 55:162–167
irradiation for painful bone metastases following single Mintz AH, Kestle J, Rathbone MP et al (1996) A randomized
fraction radiotherapy. Radiother Oncol 52:123–127 trial to assess the efficacy of surgery in addition to
Katagiri H, Takahashi M, Wakai K et al (2005) Prognostic radiotherapy in patients with a single cerebral metastasis.
factors and a scoring system for patients with skeletal Cancer 78:1470–1476
metastasis. J Bone Joint Surg Br 87:698–703 Mithal N, Needham P, Hoskin P (1994) Retreatment with
Katz AW, Carey-Simpson M, Muhs AG et al (2007) Hypo- radiotherapy for painful bone metastases. Int J Radiat Oncol
fractionated stereotactic body radiation therapy (SBRT) for Biol Phys 29:1011–1014
limited hepatic metastases. Int J Radiat Oncol Biol Phys Mornex F, Thomas L, Mohr P et al (2003) Randomized phase
67:793–798 III trial of fotemustine plus whole brain irradiation in
Kavanagh BD, Schefter TE, cardenes HR et al (2006) Interim cerebral metastases of melanoma. Cancer Radiother 7:
analysis of a prospective phase I/II trial for liver metastases. 1–8
Acta Oncol 45:848–855 Nagata Y, Negoro Y, Aoki T et al (2002) Clinical outcomes of
Khuntia D, Brown P, Li J et al (2006) Whole-brain radiother- 3D conformal hypofractionated single high-dose radiother-
apy in the management of brain metastasis. J Clin Oncol apy for one or two lung tumors using a stereotactic body
24:1295–1304 frame. Int J Radiat Oncol Biol Phys 52:1041–1046
Knisely J, Strugar J (2006) Can decompressive surgery improve Nakagawa K, Aoki Y, Tago M et al (2000) Megavoltage CT-
outcome in patients with metastatic epidural spinal-cord assisted stereotactic radiosurgery for thoracic tumors:
compression? Nat Clin Pract Oncol 3:14–15 original research in the treatment of thoracic neoplasms.
Kocher M, Sofietti R, Abacioglu U et al (2011) Adjuvant Int J Radiat Oncol Biol Phys 48:449–457
whole-brain radiotherapy versus observation after radiosur- Nielsen OS, Bentzen SM, Sandberg E et al (1998) Randomized
gery or surgical resection of one to three cerebral metas- trial of single dose versus fractionated palliative radiother-
tases: results of the EORTC 22952-26001 study. J Clin apy of bone metastases. Radiother Oncol 47:233–240
Oncol 29:134–141 Niewald M, Tkocz HJ, Abel U et al (1996) Rapid course
Koswig S, Budach V (1999) Remineralization and pain relief in radiation therapy vs. more standard treatment: a randomized
bone metastases after different radiotherapy fractions (10 trial for bone metastases. Int J Radiat Oncol Biol Phys
times 3 Gy vs. 1 time 8 Gy). A prospective study. 36:1085–1089
Strahlenther Onkol 175:500–508 Norihisa Y, Nagata Y, Takayama K et al (2008) Stereotactic
Kunkler I (2006) Surgical resection in metastatic spinal cord body radiotherapy for oligometastatic lung tumors. Int J
compression. Lancet 367:109 Radiat Oncol Biol Phys 72:398–403
572 D. Rades

Okawa T, Kita M, Goto M et al (1988) Randomized prospec- in-field recurrences of metastatic spinal cord compression.
tive clinical study of small, large and twice-a-day fraction Cancer 113:1090–1096
radiotherapy for painful bone metastases. Radiother Oncol Rades D, Kieckebusch S, Haatanen T et al (2008c) Surgical
13:99–104 resection followed by whole brain radiotherapy versus
Okunieff P, Petersen AL, Philip A et al (2006) Stereotactic whole brain radiotherapy alone for single brain metastasis.
body radiation therapy (SBRT) for lung metastases. Acta Int J Radiat Oncol Biol Phys 70:1319–1324
Oncol 45:808–817 Rades D, Kueter JD, Hornung D, et al (2008d) Comparison of
Patchell RA, Tibbs PA, Walsh JW et al (1990) A randomized stereotactic radiosurgery (SRS) alone and whole brain
trial of surgery in the treatment of single metastases of the radiotherapy (WBRT) plus a stereotactic boost
brain. N Engl J Med 322:494–500 (WBRT ? SRS) for one to three brain metastases. Strah-
Patchell RA, Tibbs PA, Regine WF et al (1998) Postoperative lenther Onkol 184:655–662
radiotherapy in the treatment of single metastases to the Rades D, Kueter JD, Veninga T et al (2009) Whole brain
brain. A randomised trial. JAMA 280:1485–1489 radiotherapy plus stereotactic radiosurgery (WBRT ? SRS)
Patchell R, Tibbs PA, Regine WF et al (2005) Direct versus surgery plus whole brain radiotherapy
decompressive surgical resection in the treatment of spinal (OP ? WBRT) for 1–3 brain metastases: results of a
cord compression caused by metastatic cancer: a random- matched pair analysis. Eur J Cancer 45:400–404
ised trial. Lancet 366:643–648 Rades D, Douglas S, Veninga T et al (2010a) Validation and
Postmus PE, Haaxma-Reiche H, Smit EF et al (2000) simplification of a score predicting survival in patients
Treatment of brain metastases of small-cell lung cancer: irradiated for metastatic spinal cord compression. Cancer
comparing teniposide and teniposide with whole brain 116:3670–3673
radiotherapy––a phase II study of the European Organiza- Rades D, Huttenlocher S, Dunst J et al (2010b) Matched pair
tion for the Research and Treatment of Lung Cancer analysis comparing surgery followed by radiotherapy and
Cooperative Group. J Clin Oncol 18:3400–3408 radiotherapy alone for metastatic spinal cord compression.
Prasad D, Schiff D (2005) Malignant spinal-cord compression. J Clin Oncol 28:3597–3604
Lancet Oncol 6:15–24 Rades D, Lange M, Veninga T et al (2011b) Final results of a
Priestman TJ, Dunn J, Brada M et al (1996) Final results of the prospective study comparing the local control of short-
Royal College of Radiologists trial comparing two different course and long-course radiotherapy for metastatic spinal
radiotherapy schedules in the treatment of cerebral metas- cord compression. Int J Radiat Oncol Biol Phys 79:524–530
tases. Clin Oncol 8:308–315 Rasmusson B, Vejborg I, Jensen AB et al (1995) Irradiation of
Rades D, Dziggel L, Haatanen T et al (2011a) Scoring systems bone metastases in breast cancer patients: a randomized
to estimate intracerebral control and survival rates of study with 1 year follow-up. Radiother Oncol 34:179–184
patients irradiated for brain metastases. Int J Radiat Oncol Roos DE, Turner SL, O’Brien PC et al (2005) Randomized trial
Biol Phys (in press) of 8 Gy in 1 versus 20 Gy in 5 fractions of radiotherapy for
Rades D, Huttenlocher S, Bajrovic A et al (2011c) Surgery neuropathic pain due to bone metastases (Trans-Tasman
followed by radiotherapy versus radiotherapy alone for Radiation Oncology Group, TROG 96.05). Radiother Oncol
metastatic spinal cord compression from unfavorable 75:54–63
tumors. Int J Radiat Oncol Biol Phys (in press) Rusthoven KE, Kavanagh BD, Burri SH et al (2009) Multi-
Rades D, Fehlauer F, Stalpers LJA et al (2004) A prospective institutional phase I/II trial of stereotactic body radiation
evaluation of two radiation schedules with 10 versus 20 therapy for lung metastases. J Clin Oncol 27:1579–1584
fractions for the treatment of metastatic spinal cord Schöggl A, Kitz K, Reddy M et al (2000) Defining the role of
compression: final results of a multi-center study. Cancer stereotactic radiosurgery versus microsurgery in the treat-
101:2687–2692 ment of single brain metastases. Acta Neurochir (Wien)
Rades D, Stalpers LJ, Veninga T et al (2005) Spinal reirradi- 142:621–626
ation after short-course RT for metastatic spinal cord Steenland E, Leer JW, van Houwelingen H et al (1999) The
compression. Int J Radiat Oncol Biol Phys 63:872–875 effect of a single fraction compared to multiple fractions on
Rades D, Stalpers LJA, Schulte R et al (2006) Defining the painful bone metastases: a global analysis of the Dutch
appropriate radiotherapy regimen for metastatic spinal cord Bone Metastasis Study. Radiother Oncol 52:101–109
compression (MSCC) in non-small cell lung cancer Sundstrom JT, Minn H, Lertola KK et al (1998) Prognosis of
(NSCLC) patients. Eur J Cancer 42:1052–1056 patients treated for intracranial metastases with whole-brain
Rades D, Schild SE, Lohynska R et al (2007a) Two radiation irradiation. Ann Med 30:296–299
regimens and prognostic factors for brain metastases in Sze WM, Shelley MD, Held I et al (2003) Palliation of
nonsmall cell lung cancer patients. Cancer 110:1077–1082 metastatic bone pain (single fraction versus multifraction
Rades D, Pluemer A, Veninga T et al (2007b) Whole-brain radiotherapy-a systematic review of randomised trials). Clin
radiotherapy versus stereotactic radiosurgery for patients in Oncol 15:345–352
recursive partitioning analysis classes 1 and 2 with 1 to 3 Timmerman RD, Bizekis CS, Pass HI et al (2009) Local
brain metastases. Cancer 110:2285–2292 surgical, ablative, and radiation treatment of metastases. Ca
Rades D, Dunst J, Schild SE (2008a) The first score predicting Cancer J Clin 59:145–170
overall survival in patients with metastatic spinal cord Tong D, Gillick L, Hendrickson FR (1982) The palliation of
compression. Cancer 112:157–161 symptomatic osseous metastases. Final results of the study
Rades D, Rudat V, Veninga T et al (2008b) Prognostic factors by the radiation therapy oncology group. Cancer 50:893–
for functional outcome and survival after re-irradiation for 899
Radiation Therapy for Metastatic Disease 573

Uematsu M, Shioda A, Tahara K et al (1998) Focal, high dose, Wu JS, Wong R, Johnston M et al (2003) Meta-analysis of
and fractionated modified stereotactic radiation therapy for dose-fractionation radiotherapy trials for the palliation of
lung carcinoma patients: a preliminary experience. Cancer painful bone metastases. Int J Radiat Oncol Biol Phys
82:1062–1070 55:594–605
Van der Linden YM, Dijkstra SP, Vonk EJ et al (2005) Wulf J, Hädinger U, Oppitz U et al (2001) Stereotactic
Prediction of survival in patients with metastases in the radiotherapy of targets in the lung and liver. Strahlenther
spinal column. Results based on a randomized trial of Onkol 177:645–655
radiotherapy. Cancer 103:320–328 Zimm S, Wampler GL, Stablein D et al (1981) Intracerebral
Vecht CJ, Haaxma-Reiche H, Noordijk EM et al (1993) metastases in solid tumor patients: natural history and
Treatment of single brain metastasis: radiotherapy alone or results of treatment. Cancer 48:384–394
combined with neurosurgery? Ann Neurol 33:583–590
Advances in Supportive and Palliative Care
for Lung Cancer Patients
Michael J. Simoff

Contents Abstract
As advancements are continuing to be made in the
1 Introduction.............................................................. 575 diagnosis and treatment of lung cancer, the number
2 Dyspnea ..................................................................... 576 of symptoms that patients present to their managing
2.1 Hypoxia...................................................................... 576 physicians with, also increases. The most significant
2.2 Chronic Obstructive Pulmonary Disease .................. 576 of these; dyspnea, cough, and hemoptysis can be
2.3 Endobronchial Disease .............................................. 577
very difficult to treat. The goal of this chapter is to
3 Pleural Disease ......................................................... 584 offer clinicians a greater awareness to the variety of
4 Tracheoesophageal Fistula...................................... 587 possible etiologies and the possible therapeutic
interventions available to them and their patients
5 Cough ........................................................................ 587
6 Hemoptysis................................................................ 588
7 Conclusion ................................................................ 589
References.......................................................................... 589
1 Introduction

The majority of patients with lung cancer will expe-


rience some symptoms (dyspnea, cough, and/or
hemoptysis) during the course of their disease. These
symptoms can greatly affect, not only the quality of
life of these patients, but may also influence the
therapeutic modalities that their physician may want
to employ to deliver further therapy.
Most physicians would define palliation as the
relief or soothing of symptoms of a disease, but not
affecting cure. The term palliation has often been
associated with end of life care of patients with can-
cer. In this chapter a broader view of palliation will be
used. As an example of this, relief of an airway
M. J. Simoff (&)
obstruction in a patient with end-stage lung cancer
Bronchoscopy and Interventional Pulmonology, may allow them not to suffocate as their cause of
Pulmonary and Critical Care Medicine, death, versus a patient diagnosed with an airway
Henry Ford Medical Center, obstruction, subsequently proven to be cancer. This
Wayne State University School of Medicine,
Detroit, MI 48202, USA
patient’s airway obstruction is relieved and the patient
e-mail: msimoff1@hfhs.org can breathe better and thereby undergo more

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 575
DOI: 10.1007/174_2011_313, Ó Springer-Verlag Berlin Heidelberg 2011
576 M. J. Simoff

aggressive therapy as well as have a diminished risk 2.1 Hypoxia


of a post obstructive pneumonia. Although ‘‘cure’’
may not be effected by the direct intervention, many When a patient is short of breath, it is often an
palliative techniques can increase survival of patients automatic response by patients, nurses and physi-
in addition to improving their quality of life. In the cians that the patient needs oxygen. Hypoxia is a
study by Brutinel et al. (1987) in a patient population common complication in patients with lung cancer.
affected by airway obstruction, 84–92% of their Hypoxia is defined as an oxygen saturation of \88%.
patients had symptomatic palliation of symptoms Some patients will have hypoxia at rest. Other
solely with laser resection of the endobronchial tumor. patients will maintain adequate oxygenation while
Survival at 7 months was better in the laser bronchos- resting but quickly desaturate with activity devel-
copy group (60%, n = 71) than in the control group oping dyspnea. Supplemental oxygen is a very
(0%, n = 25) (Brutinel et al. 1987). This idea is an common intervention to help relieve dyspnea do to
expansion on the traditional view of palliation; with desaturations in patients with hypoxia both at rest
more modern tools and techniques, this broader view and with exertion (Escalante et al. 1996). When
should be a part of all treating physicians’ thinking. patients are hypoxic with rest or with activity, the
Symptoms that patients with lung cancer may use of oxygen with sleep can often help improve
experience include: dyspnea, cough, and hemoptysis. rest. The prescription of oxygen particularly with
Many different manifestations of lung cancer (local activity should be titrated to ensure that patients are
invasion, metastasis, or paraneoplastic syndromes) receiving the appropriate doses.
may be responsible for any or all of these. The goal of Hypoxia is an objective diagnosis, but dyspnea is
this chapter will be to expand the treating physician’s not. The simple technique of directing a hand-held fan
awareness of a variety of these etiologies and a toward your face can help diminish the sense of
variety of possible therapeutic interventions. shortness of breath. Galbraith et al. (2010) have
demonstrated this in their study published in 2010,
which supports the hypothesis that a hand-held fan
2 Dyspnea directed to the face reduces the sensation of breath-
lessness. It must be remembered by practitioners that
Dyspnea will affect 65% of all patients with lung oxygen should be prescribed for patients with
cancer during some time in their disease course hypoxia, but if a patient is not hypoxic other etiolo-
(Jacox et al. 1994; World Health Organization 1990). gies of dyspnea should be looked for.
The etiologies of dyspnea can vary including:
hypoxia, progression of underlying diseases [i.e.
chronic obstructive pulmonary disease (COPD), 2.2 Chronic Obstructive Pulmonary
asthma, congestive heart failure (CHF), etc.], airway Disease
obstruction, pleural disease, deconditioning from
inactivity brought on from therapy or from extended It is not uncommon for patients with lung cancer to
stays in the hospital; depression and malnutrition can have other underlying diseases of their lungs, partic-
also be a cause as these also occur in many patients ularly chronic obstructive pulmonary disease
(Hoegler 1997). (COPD). During therapy for lung cancer, the treat-
Dyspnea is a very complex problem as the myriad ments themselves and/or infections brought on by
of etiologies or combinations of conditions can lead to immunosuppression of the treatments can lead to
a difficult time for patients and physicians. If the exacerbations of a patient’s chronic underlying dis-
etiology for the dyspnea can be identified correctly it order. Beta-2 agonists, antibiotics, and sometimes
can be managed successfully. With some type of steroid use can often improve the tracheobronchitis
palliative treatment, the patient may have a much and/or bronchospasm associated with the flare-ups.
greater tolerance for further interventions (whether it The use of aggressive treatment regimens can assist in
be radiation therapy, chemotherapy, or surgery), controlling some of the underlying lung disease,
which might have been impossible except for this which can manifest as worsening shortness of breath.
intervention. Checking airway mechanics with objective testing
Advances in Supportive and Palliative Care for Lung Cancer Patients 577

often develop worsening shortness of breath as their


therapy progresses, particularly during radiation
treatments. When radiation treatments involve the
areas of obstruction mucosal inflammation occurs
causing worsening airway obstruction. When this
becomes greater than 50%, symptoms will begin.
Therefore, endobronchial techniques should not only
be considered as the end-stage of lung cancer man-
agement but also both at the beginning and throughout
the management of patients with lung cancer (Cortese
and Edell 1993).
Lastly, when all management options have been
used, end-stage patients can develop compromise of
their airways as the cancer continues to progress. In
these situations, endobronchial techniques may ben-
efit the patient in its more traditionally accepted role
of palliation. Endobronchial management options
Fig. 1 Endobronchial obstruction from a lung cancer may help to relieve some of their symptoms,
allowing the patient freedom from shortness of
breath in conjunction with hospice or other end-
such as spirometry can assist in the assessment of of-life therapies. (Cortese and Edell 1993; Sutedja
patients with increasing shortness of breath and et al. 1995).
underlying disease. Tell-tale reductions in dynamic Most patients with dyspnea due to airways
airway volumes (FEV1 and FVC) can help guide obstruction are at home and limited but ambulatory, a
therapeutic approaches and measure response of those smaller number of patients are inpatient due to the
therapies with return to baseline values. degree of their dyspnea and others are receiving
mechanical ventilatory support due to the severity of
their disease. The majority of procedures performed
2.3 Endobronchial Disease in the United States for airway obstructions are
therefore performed on an outpatient basis. Those
Most new cases of lung cancer in the United States patients hospitalized due to dyspnea, post-obstructive
will be in an advanced stage (Cancer Facts and pneumonia, or respiratory failure, very commonly
Figures 2001). More than 50% of these patients will improve quickly, often being extubated or discharged
have some involvement of the central airways (Fig. 1) home postoperatively. This rapid symptomatic
(Luomanen and Watson 1968). This can be in the improvement allows patients to remain ambulatory
form of bulky endobronchial disease, endobronchial with an improved quality of life. It may also allow
extension, or extrinsic compression of the airways by them to continue with or begin additional anti-cancer
the tumor or by lymphadenopathy. These patients treatment. Although interventional procedures are not
may have respiratory symptoms: shortness of breath, definitive therapies, they often provide partial to total
hemoptysis, and/or cough. Many of these patients relief of the severe dyspnea produced by nearly
may benefit from endobronchial intervention as part complete airway occlusion.
of the management of their disease (Cancer Facts and Interventional pulmonary programs that include
Figures 2001). endobronchial procedures should include an arma-
We as humans have a great capacity of reserve in mentarium of therapeutic modalities rather than a
our ability to breath. It is very uncommon to have single-invasive approach to manage patients with
dyspnea until the airways are greater than 50% complicated lung cancer. As each patient’s anatomy
obstructed. Not all endobronchial disease causes differs, the manner in which the patient’s cancer leads
complete obstruction of the airways. Patients who to symptoms varies. Several procedures used in
have partial airway obstruction (less than 50%), will conjunction (i.e. laser and stenting) may be necessary to
578 M. J. Simoff

Fig. 3 Rigid bronchoscope

become the most wide-spread tool for evaluating


and diagnosing diseases of the airways and lungs
(Fig. 2) (Ikeda 1970, 1995). The rigid broncho-
scope, the flexible bronchoscope’s predecessor, was
in many regards forgotten as a tool until interven-
tional pulmonology evolved in the 1980s. Inter-
Fig. 2 Flexible bronchoscope
ventional pulmonologists reevaluated this tool and
found its properties advantageous for the procedures
provide the most effective treatment. The necessity of that are currently performed. A survey in 1991 by
multiple modalities can also be affected by the patient’s the American College of Chest Physicians reported
medical condition, for instance, patients with high- that only 8% of responding pulmonologists used a
oxygen requirements cannot be treated with traditional rigid bronchoscope (Prakash and Stubbs 1991).
thermal energy modalities. Regarding endobronchial Despite the expansion of interventional pulmonolo-
prosthesis, the use of both metallic and silastic stents is gy, the number of pulmonologists who use a
very important as each stent type has great advantages rigid bronchoscope as part of their practice remains
over the other. A program offering a wide variety of very low.
modalities allows the best selection of approaches for Overall, both the flexible bronchoscope and the
any given patient (Cortese and Edell 1993). rigid bronchoscope are necessary for the practice of
The following sections discuss a variety of tech- interventional pulmonology. The rigid bronchoscope
niques and tools available to the interventionalist. In offers many advantages to the interventional pulmo-
many cases, no one technique is better than the other, nologist, one of which is superior control of the air-
and some combination of these techniques often way. Ventilation is performed through the rigid
offers the greatest benefit to the patient. bronchoscope itself rather than around the flexible
bronchoscope. The larger-bore rigid bronchoscopes
2.3.1 Bronchoscopy allow optical systems, large caliber suction catheters,
Since the inception of flexible fiberoptic bronchos- and ablative instruments to pass through the scope
copy in the late 1960s in Japan and in 1970 in simultaneously. Large biopsy forceps are used
the United States, the flexible bronchoscope has through the rigid bronchoscope, which can provide
Advances in Supportive and Palliative Care for Lung Cancer Patients 579

more significant tissue biopsies as well as assist in


mechanical debulking of lesions.
The rigid bronchoscope itself (Fig. 3) can be used to
debulk tumor from the airway lumen. The distal end of
the bronchoscope has a beveled end. This edge can be
used to shear large sections of endobronchial tumor
away from the airway wall in a technique often referred
to as applecoring. In a report on 56 patients with
endobronchial obstruction from the trachea to the distal
mainstem bronchi, Mathisen and Grillo (1989) descri-
bed improvement in 90% of their patients. Only 3 of the
56 patients had more than minor bleeding with this
procedure. Applecoring combined with the use of lar-
ger biopsy forceps allows tumor to be quickly resected
from the obstructed airway.
There is debate in terms of the use of only a
flexible bronchoscope versus a flexible bronchoscope
in conjunction with a rigid bronchoscope. It is true Fig. 4 Main carina after laser resection of endobronchial
that more tools and techniques have been developed tumor
for the flexible bronchoscope; it is an excellent tool
for many airways procedures. The rigid broncho- The KTP laser has many of the same properties as
scope is a more difficult instrument to use than a the Nd:YAG with a delivered wavelength of 532 nm.
flexible bronchoscope, and the rigid bronchoscope The CO2 laser is more difficult to apply to endo-
requires additional training beyond the typical fel- bronchial tumors as it requires a direct line of fire and
lowship. Rigid bronchoscopy is also most commonly is therefore only used with suspension laryngoscopy.
performed in the operating room with general A new fiberoptic adaptor and fiber has become
anesthesia, limiting its availability to some pul- available from Omniguide (Omniguide Inc., Cam-
monary physicians. Despite this, to provide the best bridge, MA). Although an excellent cutting tool, the
therapeutic options for patients with airways disease, CO2 laser is not a good photocoagulation device, in
the use of both tools is necessary. contrast to Nd:YAG and YAP wavelengths and
despite the newer fiberoptic option, the CO2 laser
2.3.2 Laser Therapy remains less commonly used.
Lasers have many medical uses, including the The predominant tissue effects of lasers are ther-
endobronchial ablation of lung cancer. Several types of mal necrosis (photodessication) and photocoagulation
lasers are currently used within the bronchi: neodymium: (Fig. 4). Thermal necrosis uses higher energy levels
yttrium-aluminum-garnet (Nd:YAG), potassium- to destroy tissue, causing the formation of eschar or
titanyl-phosphate (KTP), and carbon dioxide (CO2). actual photodessication of tissue. This technique must
Newer diode as well as the yttrium–aluminum– be used cautiously as most lung cancers have signif-
perovskite (YAP) lasers have also entered the mar- icant vascularity. When destroying tissue with laser
ketplace. Still, the most commonly used endoscopic energy, large blood vessels can be perforated with the
laser is the Nd:YAG, which delivers energy at tissue destruction, leading to significant hemorrhage.
a wavelength of 1,064 nm. The laser energy is The technique of photodessication is an excellent tool
conducted via a quartz monofilament and thus can be for managing airways disease, but does require a
easily used with either the rigid or flexible broncho- skillful operator. Photocoagulation uses lower energy
scopes. Normally, Nd:YAG is used at 30–60 W, but it levels with longer exposure intervals, causing tumors
has a wide range of power outputs, up to 100 W. to ‘‘shrink’’ and diminishing blood flow to that region.
Depending on the energy level used, the laser can By devascularizing the tumor, more rapid mechanical
treat superficially, penetrate tissue several millimeter debulking can be performed with improved control of
in depth, or provide complete tissue destruction. bleeding.
580 M. J. Simoff

Table 1 Advantages/disadvantages of silastic stents


Advantages
Removable and replaceable
No growth through stent
Low cost
Low likelihood of granulation tissue formation
Disadvantages
Potential for migration/dislodgment
Rigid bronchoscopy needed for placement
Possible secretion adherence

Table 2 Advantages/Disadvantages of Metal Stents Fig. 5 Montgomery T-tube

Advantages
Easy to place to improve angulations or exposure of parts of an
Good wall/internal diameter relationship airway obstruction.
Powerful radial force The reported success rate of symptom palliation
Excellent conformity for irregular tracheal or bronchial using laser energy in the endobronchial management
walls of lung cancer is high. Reports of clinical improve-
Good epithelialization ment rates range from 84 to 92% following laser
Disadvantages bronchoscopy (Dumon et al. 1982; Beamis et al.
1991; Cavaliere et al. 1988; Kvale et al. 1985;
Permanent
Eichenhorn et al. 1986). Other studies demonstrate
Tumor regrowth (non-covered)
improved survival in patients treated with laser
Possible migration of covered stents bronchoscopy (Brutinel et al. 1987; Desai et al. 1988;
Significant granulation tissue stimulation Stanopoulos et al. 1993; Petrovich et al. 1981).
Epithelialization adversely affecting wall mechanics and
secretion clearance 2.3.3 Endobronchial Prosthesis
Radial force causing necrosis of bronchial wall, erosion, Endobronchial prosthesis are airway stents, which can
fistulas, perforation
be placed in response to several clinical situations:
intrinsic, extrinsic, or mixed endobronchial obstruc-
tion. Stents are composed of silastic rubber, metal
Laser therapy can be performed via either flexible alloys, or hybrids (mixed materials). Stents work well
or rigid bronchoscopy. Many interventionalists prefer in conjunction with other modalities such as laser and
rigid bronchoscopy for laser procedures when possi- mechanical debulking of tumors. Advantages and
ble. Nd:YAG laser fibers can be passed through the disadvantages of each stent type are noted in Tables 1
working channel of most flexible bronchoscopes. An and 2.
advantage of using the flexible bronchoscope is that
laser energy can be delivered to areas that cannot be 2.3.3.1 Silastic Stents
reached with a rigid bronchoscope (Brutinel et al. Many of the silastic stents now in use evolved from
1987; Mathisen and Grillo 1989; Hetzel et al. 1983; the Montgomery T-tube, which was first used in the
Mehta et al. 1985; McDougall and Corese 1983; Toty early 1960s. This T-shaped stent supports the entire
et al. 1981; Dumon et al. 1982; Arabian and Spagnolo trachea with an arm that extends through a tracheot-
1984; Beamis et al. 1991; Sonett et al. 1995; Macha omy. In patients with a patent tracheostomy, the
et al. 1994; Desai et al. 1988; Stanopoulos et al. 1993; Montgomery T-tube remains an excellent tool for the
Cavaliere et al. 1994; Ross et al. 1990). For this management of endotracheal, particularly sub-glottic
reason, a fiberoptic bronchoscope can be inserted disease (Fig. 5) (Colt and Dumon 1993; Cooper et al.
through the rigid bronchoscope whenever necessary 1989; Montgomery 1965).
Advances in Supportive and Palliative Care for Lung Cancer Patients 581

Fig. 6 a The Dumon silastic


stent. b The Dumon-Y stent

In 1990, Dumon (1990) reported the use of what is


now referred to as the Dumon stent (Novatech, Plan
de Grasse, France). Developed in 1987, it is a silastic
stent with evenly spaced studs along its outside walls.
(Fig. 6a, b) The studs are intended to minimize
migration of the stent in the airway. The studs also
allow the clearance of secretions around the walls of
the stent.
Dumon stents are effective in maintaining their
structural integrity when placed endobronchially. The
solid walls of the stent prevent tumor growth from
re-obstructing airways. In the situation of a newly
diagnosed lung cancer with airways obstruction, the
endobronchial tumor can be debulked and then a stent is Fig. 7 The Hood bronchial stent
placed prior to the initiation of radiotherapy, chemo-
therapy, or both. Both external beam radiotherapy and
brachytherapy can be used with a Dumon stent in place. Another silastic stent is the hood stent (Hood
Another advantage of the Dumon stent is the ease of Laboratories, Decatur, Georgia). The Hood stent is
its removal. This can be important when endobronchial similar to the Dumon stent in design and use. The
procedures are used early in the management of cancer Hood stent is placed in the same manner as the
patients. After definitive therapies have been used Dumon stent, using a rigid bronchoscope (Fig. 7)
(radiation, chemotherapy), re-evaluation of the airway (Gaer et al. 1992).
can be performed, at which time the stent can be left in
place, removed (if deemed of no further clinical 2.3.3.2 Metallic Stents
advantage), or replaced with a larger stent that would Metallic stents, such as the ultraflex (Boston Scien-
further improve the caliber and stability of the airway. tific, Boston, MA) and AERO stent (Merit Medical
The disadvantages of the Dumon stent are the potential Systems, Inc., South Jordan, Utah) have been used in
for migration and the need for a rigid bronchoscope the endobronchial management of lung cancer. The
for placement. Migration occurs less often when an advantage of metal stents is the relative ease for
experienced interventional endoscopist places the stent placement via a flexible bronchoscope with fluoro-
(Petrovich et al. 1981; Dumon 1990; Mehta 2008; scopic assistance. This ease of placement has led
Diaz-Jimenez et al. 1994; Freitag et al. 1995; Clarke some bronchoscopists to use these stents as their only
et al. 1994). method to manage endobronchial disease. Such a
582 M. J. Simoff

Fig. 9 The AERO stent

Fig. 8 The Ultraflex stent-covered and uncovered

practice limits the options to patients who may


otherwise be available if all interventional modalities
were offered. The wire mesh design of many of the
original metal stents did not prevent the tumor from
growing through the stent. The ultraflex stent is also
available in covered versions and the AERO stent is
covered. A wrap is applied to the outside of the wire
mesh to prevent tumor invasion through the stent. Fig. 10 The Dynamic-Y hybrid stent
There is good data for the use of these stents in the
endobronchial management of lung cancer (Colt and epithelization changes the mechanics of the airways
Dumon 1991, 1993; Mehta 2008; Bolliger et al. 1993; with time by making them stiffer, which may lead to
Gelb et al. 1992). further airway complications (Freitag et al. 1995;
Ultraflex stents are made of nitinol, a titanium and Gelb et al. 1992). This may not be a concern in situ-
nickel alloy, which has little bioreactivity. This stent ations of palliation of late-stage disease, but must be
has excellent inner to outer diameter ratio and con- considered if long-term survival is expected. Another
forms well to various airway shapes, maintaining an consideration with metal stents is that once they are
equal pressure along the entire length of the stent inserted, their removal can be difficult and often
despite expansion size. Ultraflex stents are available impossible. Although uncommon, another risk with
in a variety of lengths and diameters. Overall the the use of metal stents is the erosion that can occur
covered version of this stent is excellent for use in through bronchial/tracheal walls. This was more of a
palliation of airway obstruction (Fig. 8). concern with the older Gianturco stents, which are no
AERO stents (previously alveolus stents) are longer used, than with newer metallic stents.
another metallic stent, which is available for clinical
use (Fig. 9). This stent has similar advantages as the 2.3.3.3 Hybrid Stents
Ultraflex stents, but are completely covered form end- The Dynamic-Y stent (Boston Scientific, Boston,
to-end. This added covering can assist intervention- MA) is a hybrid stent with silastic walls and stainless
alists in limiting tumor from reinvading airways after steel c-rings that artificially represent the cartilage
resection. It has also been demonstrated that the (Fig. 10). The posterior wall of the stent is made of a
AERO stents complete coverage allows less compli- thinner silastic rubber to make it more dynamic,
cated removal, much like the Dumon stent (Dumon similar to the membranous trachea. The three avail-
1990). able sizes of this stent are designed to traverse the
The uncovered portions of metal stents epithelial- entire length of the trachea with branches into the
ize as they remain in the airways, thereby becoming right and left mainstem bronchi. The Dynamic-Y stent
incorporated into the wall of the bronchus. This requires rigid bronchoscopy and is difficult to place,
Advances in Supportive and Palliative Care for Lung Cancer Patients 583

sodium. The laser energy is transmitted via a flexible


quartz fiber, which can be used through either a
flexible or rigid bronchoscope. The fiber tip can be
placed in close proximity to the tumor mass or it can
be imbedded into the tumor to provide the energy
needed to start the intracellular activation of the
porfimer sodium. This reaction leads to an intracel-
lular photooxidative reaction and subsequent cellular
Fig. 11 The Polyflex stent destruction. Tissue necrosis ensues as the cancer cells
die (Furuse et al. 1993; Hayata et al. 1993; Moghissi
et al. 1999).
remaining uncommon in clinical practice. Despite As the neoplastic tissue becomes necrotic, it must
this, the Dynamic-Y stent offers excellent results be removed. This requires repeated bronchoscopies,
when placed in the appropriate. the first within 36 h of the treatment. Bronchoscopy
The Polyflex stent (Boston Scientific, Boston, MA) can then be repeated as it is deemed necessary by the
is another hybrid stent made of woven polyester degree of necrosis and tissue sloughing identified.
monofilament network with a complete coating of One of the reasons PDT is a poor selection for
silicone. The Polyflex stents solid wall design pre- obstructing airways disease is that the necrosis of
vents tumor growth but this stent does have a very bulky tumor can be dangerous to the patient if the
high expansion pressure to the airway wall (Fig. 11). necrotic tissue separates from the bronchial wall and
Stents are effective tools for the endobronchial occludes the airway. In programs that use only PDT
management of lung cancer. Stents should be chosen for obstructing airway tumors, patients remain intu-
carefully, weighing advantages and disadvantages of bated following the procedure for 1–2 days because
each. of this concern. If necrotic tissue is removed over the
first 24–48 h, a second laser application to the cancer
2.3.4 Photodynamic Therapy can be performed, thus improving the cancer tissue
Photodynamic therapy (PDT) is an adjunctive destruction.
modality to the management of endobronchial dis- PDT is an excellent therapeutic modality for
ease. It has been reported in use with bulky patients with early-stage cancers. It destroys neo-
obstructing airways disease, but it has a limited role plastic tissue effectively and is an outstanding thera-
and does not replace Nd:YAG lasers, stents, and rigid peutic modality in the management of carcinoma in
bronchoscopy (Lam 1994; Sutedja et al. 1994). The situ and microinvasive cancers. Further discussion of
most suitable lesions for PDT are in situ carcinomas this is beyond the scope of this chapter. PDT is a
or those limited to 2–4mm of microinvasion (not necessary tool in the armamentarium of endobron-
through the cartilaginous layer of the airway) (Furuse chial treatments, but the time delays and multiple
et al. 1993). steps of management make it a more cumbersome
A photosensitizing drug is intravenously adminis- therapy for the management of late-stage endobron-
tered to the patient 48–72 h prior to the procedure. chial lung cancer (Moghissi et al. 1999).
Porfimer sodium (Photofrin, Axcan Pharma Inc.,–
United States, Birmingham, Alabama) is the most 2.3.5 Cryotherapy
common agent used. This photosensitizer penetrates Cryotherapy is another method to destroy malignant
all cells systemically. It is not cleared as quickly from tissue that obstructs the tracheobronchial tree. Tissue
cancer cells as in most other cells of the body and is is frozen and then thawed to destroy it, instead of the
therefore found in higher concentrations in cancer heat used in laser-based technologies. A probe is
cells as opposed to the endothelium surrounding the placed onto or into an obstructing tumor mass. Liquid
tumor at the time of treatment (Furuse et al. 1993; nitrogen (–196°C) or nitrous oxide (–80°C) cools
Hayata et al. 1993). An argon dye or diode laser is the probe tip when performing cryotherapy. The
then used to provide the 632 nm wavelength light tissue freezing and thawing cycle used in cryotherapy
energy required to activate the intracellular porfimer leads to the destruction of all cells in an area of
584 M. J. Simoff

approximately 1 cm in diameter of the probe tip. causing a similar thermal effect as electrocautery. This
The vascular thrombosis that occurs with the super- delivery of energy allows large areas to be treated
cooling of tissue minimizes the bleeding during the relatively quickly and can be an ideal tool when
eventual resection of the tumor. significant bleeding is encountered. On the other hand,
The limiting factor to using cryotherapy is that the more defined area of contact with the electrocau-
the tissues destroyed with the freezing procedure take tery delivery devices allows a higher energy to be
time to die and necrose. This requires another delivered point specific to the tissue, creating an
procedure to remove the necrosed tissue and, in some excellent tool for cutting as well as coagulating.
cases, repeating treatments. Although cryotherapy is
effective at tumor destruction and management, the 2.3.7 Balloon Dilatation
necessity of repeated procedures makes this a more Balloons used for intravascular procedures can be
time-consuming technique to perform, limiting its used to manage endobronchial stenosis secondary to
usefulness in the management of bulky endobronchial both malignant and benign disease. Most balloons
disease causing severe dyspnea (Maiwand and come in a variety of diameters and lengths to help
Homasson 1995). An advantage of cryotherapy is that dilate areas of bronchial compromise. Some currently
it can be used at any level of oxygen (FiO2) a patient available balloons will dilate to three different
may require to correct hypoxia. Laser, electrocautery, diameters depending on the pressure applied to the
and argon plasma coagulation all must be used in an balloon. Malignant strictures are sometimes dilated
environment with an FiO2 \40%. If the FiO2 is prior to the placement of a stent or even used inside of
greater than 40%, the risk of an airway fire becomes a stent to fully expand it once it has been placed.
very high and places the patient at significant risk. The balloon is passed endobronchially via either a
rigid or flexible bronchoscope. The appropriate
2.3.6 Electrocautery/Argon Plasma diameter and length of the balloon are chosen for the
Coagulation particular lesion. Ideally, 5–10 mm of balloon should
Electrocautery devices or the argon plasma coagula- extend beyond the lesion both proximally and distally.
tion (APC) catheters can be introduced through a The treatment should be performed as a series of
flexible bronchoscope (one that is grounded and dilatations with gradual increase in the balloon
designed for this therapy) and can then be used to diameter to minimize the risk of tracheobronchial
debulk endobronchial disease. With both devices rupture. The balloon is inflated with a fluid, usually
electrical energy is used to cauterize tissue, thus saline. The use of fluid provides a even more distri-
minimizing the bleeding that occurs with tumor bution of pressure across the entire balloon rather than
resection. Endobronchial electrocautery treatment can the unequal pressures seen when air is used to inflate
be used similar to laser therapy and/or cryotherapy for the balloon. Once inflated to the prescribed pressure,
managing advanced endobronchial lung cancer the dilatation pressure should be maintained for
(Gerasin and Shafirovsky 1988). 1–2 min. Two minutes is preferable if the patient can
Electrocautery uses unipolar electrodes to deliver tolerate this without discomfort or hypoxia.
electric current to the tissue. The delivered energy Balloon dilatation is an adjunctive therapy to
affects the tissue in three ways: an electrolytic effect bronchoscopy, laser, and/or stenting. When used
(altering chemical bonding), a capacitance effect alone, its effects are most often temporary and
(affecting the electrical potential of local structures), symptom recurrence sill often occurs.
and a thermal effect (due to the resistance of the tissue
to he flow of electrical current). Of these, the thermal
effect is clinically that, which is most desired. 3 Pleural Disease
Argon plasma coagulation, instead of using a uni-
polar contact delivery mechanism for electrical Malignant pleural effusions occur in 7 to 15% of lung
energy, uses ionized argon gas as the conductance cancer patients (Cohen and Hossain 1966; Emerson
medium between the electrode and tissue. This non- et al. 1959; Johnston 1985; Le Roux 1968), greater than
contact tool allows a ‘‘painting’’ of the desired area half of whom develop dyspnea (Chernow and Sahn
with, in essence, a gaseous form of electrical energy 1977). The mechanism of dyspnea with pleural
Advances in Supportive and Palliative Care for Lung Cancer Patients 585

effusions is unclear. Mechanical factors influencing the pleural biopsy are performed greater than 20% of
chest wall, mediastinum, pleural space, and lung itself effusions remain undiagnosed (Storey et al. 1976;
may all contribute to the sensation of dyspnea in the Hirsch et al. 1979; Lamy et al. 1980).
patient with a pleural effusion. In institutions where medical thoracoscopy is not
Pleural effusions in the setting of lung cancer may performed, most thoracic surgeons can perform
be malignant or benign. The primary techniques used video-assisted thoracoscopic surgery (VATS). VATS
to diagnose malignant pleural effusions are: thora- differs from medical thoracoscopy in that patients
centesis, closed needle pleural biopsy, and pleuros- undergoing VATS will have general rather than
copy or medical thoracoscopy. moderate anesthesia, they will be intubated, usually
Thoracentesis is the most common technique used with a double lumen tube, rather than be spontane-
in the initial evaluation of pleural effusion. Pleural ously breathing, and three incisions will be made
fluid obtained by thoracentesis yields cytology posi- rather than the typical one (two for certain proce-
tive for malignant cells in 62–90% of true malignant dures). VATS is an excellent procedure, but is very
pleural effusions (Hsu 1987; van de Molengraft and aggressive for a diagnostic procedure in an otherwise
Vooijs 1988; Starr and Sherman 1991; Loddenkemper uncomplicated pleural effusion.
et al. 1983). Closed-needle pleural biopsies remain an The major indication for treating a pleural effusion
option for the evaluation of a malignant pleural is for the relief of dyspnea. Once the diagnosis has
effusion. Pleural biopsy historically has a lower been made, a therapeutic plan needs to be established;
diagnostic yield than cytology from thoracentesis, remembering that the etiology of the dyspnea is more
40–75% (Starr and Sherman 1991; Loddenkemper complex than the amount of fluid identified in the
et al. 1983; Prakash and Reiman 1985; Poe et al. pleural space (Estenne et al. 1983; Light et al. 1986;
1984; Escudero Bueno et al. 1990). There is a 7–12% Agusti et al. 1997; Karetzky et al. 1978; Brown et al.
additive yield from closed needle biopsy over cytol- 1978; Krell and Rodarte 1985), and may be related to
ogy alone (Starr and Sherman 1991; Loddenkemper problems with the lung itself (lymphangitic spread of
et al. 1983; Prakash and Reiman 1985). Perhaps tumor, atelectasis, direct tumor invasion, etc.). The
because of this small, added benefit, the practice of limitation of diaphragmatic movement caused by fluid
closed-needle pleural biopsies has diminished in most accumulation is a major mechanism of dyspnea in
clinical practises. patients with untreated pleural effusions. Trapped
Medical thoracoscopy is a procedure more com- lung due to parenchymal or pleural disease will
monly being used by non-surgeons for the diagnosis minimize the relief of dyspnea by the evacuation of
and treatment of pleural effusions. This technique has pleural fluid and/or pleurodesis. Therefore initially, a
excellent results in the diagnosis and treatment of therapeutic thoracentesis should be performed to
malignant pleural effusions in appropriate popula- assess the effects upon breathlessness by fluid
tions. In a study of patients being evaluated for removal and the ability of the lung to re-expand, as
malignant effusion, all enrolled patients had cytologic well as the rate and degree of reaccumulation. Chest
assessment by thoracentesis, closed-needle pleural X-ray should be performed pre- and post-therapeutic
biopsies, followed by thoracoscopy. This representa- thoracentesis to evaluate for lung re-expansion.
tive study demonstrated diagnostic yields of 62% for Chest radiographs should be used to assess as to
thoracentesis, 44% for closed-needle pleural biopsy, whether or not the pleural fluid is free flowing or loc-
with a combined sensitivity of 74%, and a diagnostic ulated, as well as the mediastinal position in respect to
yield for medical thoracoscopy of 95% (Loddenkemper the volume of the pleural effusion. Contralateral shift of
1998). Other studies have demonstrated similar the mediastinum with large effusions suggests that
results (Boutin et al. 1981; Oldenburg and Newhouse evacuation of the effusion should provide relief of
1979; Menzies and Charbonneau 1991; Canto et al. dyspnea to the patient. Expert opinion would suggest
1977). After medical thoracoscopy had been per- that no greater than one to one and a half liters of
formed less than 10% of effusions remain undiag- effusion be removed at each thoracentesis, stopping
nosed (Boutin et al. 1981; Canto et al. 1977; earlier should the patient experience dyspnea, chest
Loddenkemper 1981; Martensson et al. 1985), while pain, or coughing. The coughing and/or pain experi-
after thoracentesis for cytology and closed-needle enced by a patient is considered to be due to the
586 M. J. Simoff

expansion of the lung. It is suggested that this instillation, or during medical thoracoscopy or video-
subpopulation of patients (those that have pain, etc.) assisted thoracic surgery, with talc poudrage.
may benefit from immediate chest tube placement with Poudrage and slurry pleurodesis methods demon-
pleural evacuation or medical thoracoscopy with strated clinical success rates of 91% with no signifi-
pleurodesis due to the common belief that the patient’s cant difference in recurrence rates of effusions
lung is re-expanding (ATS Guidelines 2000). (Hartman et al. 1993; Hamed et al. 1989; Fentiman
Ipsilateral or at least no contralateral mediastinal et al. 1986; Kennedy et al. 1994; Todd et al. 1980;
shift identified on chest radiographs suggests trapped Fentiman et al. 1983; Yin et al. 1996). The greatest
lung or endobronchial obstruction, potentially limiting concern with the use of talc is the 1% risk of devel-
the relief of dyspnea a patient may experience with oping fatal acute respiratory distress syndrome
evacuation of pleural fluid. Limited removal of fluid (ARDS) and the 4% risk of non-fatal ARDS reported
(\300 ml) by thoracentesis is suggested in this sub- in the literature. Most of the ARDS complications
population to minimize reducing the pleural pressure have been reported in the United States. It has been
rapidly and increasing the risk of re-expansion pul- suggested that this may be due to the size of the talc
monary edema in these patients (ATS Guidelines 2000). particles used here versus those used in Europe
Pleural pressure monitoring can be performed (Milanez Campos et al. 1997; Rehse et al. 1996).
before, during, and after thoracentesis to determine Despite these reported risks, talc is the most com-
the amount of fluid that can be removed in a physi- monly used pleurodesis agent.
ologic manner. The use of this technique may mini- Other pleurodesis agents used include doxycycline,
mize the risk of re-expansion pulmonary edema and which when compared to historical controls had a
help assess for the presence of a trapped lung at the similar clinical success rate as previous studies with
time of the diagnostic/therapeutic thoracentesis tetracycline, 80–85% (Patz et al. 1998; Heffner et al.
(Rodriguez-Panadero and Lopez-Mejias 1989; Light 1994; Pulsiripunya et al. 1996). Bleomycin has been
et al. 1980; Lan et al. 1997). Pleural pressure moni- used and compared in randomized testing to tetracy-
toring may be a more objective assessment for trap- cline, and found to have similar complete response
ped lung than chest radiograph assessment but is rates (Hartman et al. 1993; Moffett and Ruckdeschel
complex and not regularly practiced. 1992; Martinez-Moragon et al. 1997). Doxycycline
Therapeutic modalities for managing malignant when compared directly with bleomycin had a 79%
pleural effusions include repeated therapeutic thora- complete response to bleomycin’s 72% (Hayata et al.
centesis, chemical pleurodesis via chest tube or medical 1993). When bleomycin was compared to talc, talc
thoracoscopy, pleuroperitoneal shunting, pleural demonstrated superior complete response rates in all
drainage catheters, and systemic therapy. Repeated studies (Walker-Renard et al. 1994; Hamed et al.
therapeutic thoracentesises are a viable option for those 1989; Zimmer et al. 1997).
patients with poor performance status or with advanced The use of pleuroperitoneal shunting has been
disease. There are no studies upon which to base reported for the management of malignant and other
repeated thoracentesis. If the malignant pleural effusion intractable pleural effusions. All of these studies are
continues to accumulate, a more definitive procedure case series rather than randomized in any fashion.
can be considered. A variety of new and old agents can Initial data looks promising, but it has not been eval-
and are being used for pleurodesis. uated in head-to-head studies with more conventional
Chemical pleurodesis has reported a complete treatment methods (i.e. chest tube drainage with
response rate of 64%. A comprehensive review of chemical pleurodesis) (Ponn et al. 1991; Pope and
pleurodesis further discussed response; fibrosing Joseph 1989; Schulze et al. 2001; Reich et al. 1993;
agents as a group had a 75% complete response, with Petrou et al. 1995).
talc specifically, 93%. Antineoplastic agents had a Another technique, tunneled long-term catheter
reported complete response at initial pleurodesis of drainage of the pleural space is also found in several
44% (Walker-Renard et al. 1994). studies in case series formats. These studies suggest
Talc is currently the sclerotic agent of choice for good results for the relief of dyspnea over extended
pleurodesis and can be used either via chest tube time in patients with malignant effusions. Although
placement with pleural evacuation and talc slurry encouraging, many of these studies are retrospective
Advances in Supportive and Palliative Care for Lung Cancer Patients 587

in assessment with no comparison to other treatment of the lung, and ensure the patient receives nutrition and
modalities (Chen et al. 2000; Pien et al. 2001; Pollak fluid. By addressing all of these issues, the most debili-
et al. 2001). One device, the PleurX catheter (Denver tating symptoms of this condition, the dyspnea and
Biomedical, Golden, CO) is an important addition to coughing, are also corrected.
the management of malignant pleural effusions. This Double stenting of the tracheo-bronchial tree and
device when placed into the pleural space, allows the the esophagus appears to be the procedure that yields
patient to drain a portion of their pleural effusion on a the best overall results for symptomatic relief in
daily basis, thereby controlling the build-up of fluid patients with this condition. Clinical series have
and in doing so, limiting the dyspnea patients expe- attempted either esophageal or tracheo-bronchial
rience due to this complication. When used daily, one stenting individually with mixed results. Most series
study (Putnam et al. 1999) suggests that approxi- with higher success rates use a double-stenting tech-
mately 50% of these patients will experience pleu- nique. With limited published information, our clini-
rodesis without the use of sclerotic agents in a median cal experience has been most successful with initial
of 25 days. Such techniques should be explored bronchial stenting followed in close succession with
further to fully understand their possible palliative esophageal stenting (Freitag et al. 1996; Colt et al.
implications. 1992; Alexiou et al. 1998; Koeda et al. 1997; Spivak
For malignant effusions due to small-cell lung et al. 1996; Cook and Dehn 1996).
cancer the lung cancer therapy of choice is systemic Placement of a percutaneous entero-gastric (PEG)
chemotherapy. Often these patients will respond or percutaneous entero-jejunal (PEJ) tubes can ensure
with resolution of pleural effusions and dyspnea proper nutrition and fluid management in patients
(Livingston et al. 1982). with tracheoesophageal fistulas. Patients may be able
to eat once the double stenting is performed, but
maintaining adequacy of fluid status and nutrition is
4 Tracheoesophageal Fistula often difficult.

Tracheo-esophageal fistulas are serious complications


of lung and esophageal cancer. The life expectancy 5 Cough
after the development of a tracheoesophageal fistula
with no therapy is estimated as 1–7 weeks. Patients Cough can be a debilitating symptom for some
have repeated aspiration of food, gastric contents, and patients with lung cancer. As with dyspnea, the eti-
saliva. This persistent aspiration leads to patient ology of the cough should be identified to best treat a
distress due to coughing and shortness of breath. patient. Cough can be a manifestation of endobron-
Patients can develop recurrent pneumonia with per- chial disease, pleural disease, or tracheoesophageal
sistent inflammation of the airways. Patients frequently fistula as discussed above. Cough can also originate
lose weight and become dehydrated secondary to their from something as uncommon as endobronchial irri-
intolerance of taking anything by mouth. Even with tation status-post resection, when the staples migrate
abstinence from eating and drinking most patients endobronchially and become foreign bodies in the
continue to have symptoms due to lack of control of airways. Or cough may be a manifestation of some-
their own secretions and reflux of gastric contents. thing more common, such as the patients underlying
Curative resection of the involved tracheal–bronchial COPD with or without a tracheobronchitis. Again, the
and/or esophageal segments in face of a malignancy most useful management remains that specifically
should not be considered, as most of these patients are at suited to the patient’s individual problem.
the end-stage of their lung cancer and palliative man- Sometimes though, the etiology of cough is never
agement should be emphasized. Esophageal bypass identified. It is in these situations where cough
procedures should also not be considered, as they have suppressants like benzonate (Doona and Walsh 1998)
very high morbidity. or opiates (particularly codeine) can be used.
The goals of therapy for tracheoesophageal fistula are Occasionally beta-2 agonists are prescribed with
to restore patency of the trachea, bronchi, and/or identified underlying COPD but are only occasionally
esophagus, to prevent spillage and further contamination of significant benefit (Kvale et al. 2003).
588 M. J. Simoff

hemoptysis (Cahill and Ingbar 1994; Jean-Baptiste


6 Hemoptysis 2000). Reversal of any coagulation disorders should
to be considered at the time of hemodynamic man-
Hemoptysis will be the presenting symptom in 7–10% agement. If the bleeding site is known, the bleeding
of lung cancer patients. About 20% of lung cancer lung should be placed in the dependent position to
patients will have hemoptysis some time during their help protect the non-bleeding lung. Cough suppres-
clinical course, with 3% having terminal massive sion with a narcotic (particularly codeine) can be used
hemoptysis (Miller and McGregor 1980; Chute et al. to help minimize further endobronchial bleeding in
1985; Hyde and Hyde 1974; Grippi 1990; Frost et al. non-intubated patients.
1984). Massive hemoptysis, that which most com- Bronchoscopy is often used to identify the source of
monly requires intervention, has a broad definition as bleeding. Early bronchoscopy to assess the site of
expectoration of from 100 to 600 ml of blood in 24 h. bleeding is recommended. Studies have demonstrated
Blood clot formation obstructing airways is suggested identification of the bleeding site 91% of the time when
as the most common cause of respiratory insufficiency performed early versus 50% when delayed (Credle
from massive hemoptysis. et al. 1974). A more recent retrospective study had
Initial evaluation of patients with known lung much less supportive results, with the limitation of
cancer in a specific location is somewhat different early being defined as less than 48 h. Despite this, these
from that of those patients without a known diagnosis. results suggest early bronchoscopy which is indicated
Massive hemoptysis due to lung cancer has a much (Gong et al. 1981). The goal of early bronchoscopy
poorer prognosis than hemoptysis of other etiologies. should first be to lateralize the bleeding side, secondly
One retrospective review defined the mortality of localization of the specific site to a lesion, lobe, or
massive hemoptysis as 59% in patients with bron- segment, and lastly identify the lesion that is bleeding
chogenic carcinoma (Corey and Hla 1987). In many whenever possible.
of these patients surgery, a more definitive therapeutic In the patient with hemoptysis, several studies have
modality is not on the algorithm for intervention in looked at the use of early high-resolution computed
that many of these patients are already non-surgical tomography (HRCT). In those patients without a
candidates from their primary disease. diagnosis, this technique appears to have benefits. The
The initial priority in managing a patient with use of HRCT may help diagnose: bronchiectasis, an
massive hemoptysis should be, maintaining adequate aspergilloma, and possibly identify a previously undi-
airway protection (Cahill and Ingbar 1994; agnosed lung cancer (Set et al. 1993; McGuiness et al.
Jean-Baptiste 2000). This may require endotracheal 1994; Muller 1994; Hirschberg et al. 1997). In the
intubation to maintain good control. It is suggested patient with the known diagnosis of lung cancer, this
that use of a single lumen endotracheal tube is of technique will be of limited value, particularly in those
greater benefit than double-lumen endotracheal tubes patients who have had previous radiation therapy.
(Strange 1991). A very important therapeutic tool in the manage-
Standard endotracheal intubation should use the ment of hemoptysis due to malignancy is external
largest tube possible. Occasionally selective right or beam radiation (Hoegter 1997). Prior to initiation of
left mainstem intubations are performed to protect the this therapy, other procedures are often necessary to
non-bleeding lung. This technique can be beneficial in temporize the patient and localize the area that needs
protecting the good lung, but the fact that when a right to be treated.
sided intubation is performed, it often occludes the Endobronchial management of hemoptysis should
right upper lobe and the difficulty of selective left be subdivided into identification of the location of the
sided intubations need to be considered prior to bleed (i.e. bleeding from the anterior segment of the
attempting this. left upper lobe) versus bleeding from an identified
Optimization of oxygenation needs to then be source (i.e. bleeding from an endobronchial tumor).
undertaken to clinically stabilize the patient with When the location of the bleed is identified, but no
massive hemoptysis. Next, assessment and manage- direct source is found, endobronchial management
ment of cardiovascular/hemodynamic status has to includes: bronchoscopic tamponade of the segment,
take place for proper management of the patient with usually recommended with continuous suctioning
Advances in Supportive and Palliative Care for Lung Cancer Patients 589

collapsing the segment (Zavala 1976). The use of previously not amenable to surgical treatment, the
vasoactive drugs (i.e. 1:10,000 epinephrine solution) addition of hemoptysis to this scenario should not
is suggested, although this is most useful on visual- give cause to surgical intervention at the time of this
ized lesions (Magee and Williams 1982; Worth et al. complication. In the case where a cancer is newly
1987). Ice saline lavage is discussed as a temporizing diagnosed at the time of management of hemoptysis,
technique for control of hemoptysis (Sahebjami 1976; controlling the hemoptysis with other techniques to
Conlan and Hurwitz 1983). Balloon tamponade allow full assessment/staging prior to acute surgical
techniques, using a variety of different balloons, can management should be performed. Rarely, in a life-
control hemoptysis and minimize risk of further threatening situation, surgical intervention for both
aspiration of blood. It is suggested that balloons the hemoptysis and the lung cancer may be effective.
remain in place for 24–48 h to allow tamponade of Extreme measures are sometimes required in cases
hemoptysis (Schlehe et al. 1984; Tsukamoto et al. of massive hemoptysis. Our case report was due to the
1989; Bense 1990). erosion of the pulmonary artery into the right main-
When an endobronchial source of bleeding is iden- stem bronchus with massive persistent bleeding. After
tified, attempts with vasoactive drugs can be used, but stabilizing the patient, we had the entire right pul-
often this type of bleeding requires a more aggressive monary artery embolized, through a complex proce-
mode of management. Use of Nd:YAG photocoagula- dure. This completely removed blood flow and
tion is an efficient tool for the management of bleeding perfusion to the right lung. As this would put a huge
endobronchial lesions with a reported response rate of physiologic burden on the patient, we next filled the
60% (Hetzel and Smith 1991; Jain et al. 1985; Clarke airways of the right lung with fibrin glue and placed a
et al. 1994). Use of electrocautery is also suggested in stent over the airway, eliminating all ventilation.
the literature but support other than anecdotal reporting After this was accomplished, the patient was then
is limited for the management of hemoptysis. Use of extubated and was soon ambulatory on two liters
argon plasma coagulation in one study demonstrated nasal cannula oxygen. He was home until his death
resolution of hemoptysis in 100% of patients with a 9 months later of different complications of his lung
3 month follow-up (Morice et al. 2001). cancer (Chawla et al. 2009).
Bronchial artery embolization appears to be a
semi-definitive therapy for hemoptysis. Embolization
stops bleeding in greater than 85% of all patients for 7 Conclusion
whom it is used. This excellent success rate should be
tempered with the fact that 10–20% of these patients There are many symptoms associated with lung can-
have rebleeding in the next 6–12 months (Mal et al. cer that can be palliated to allow patients the oppor-
1999; White 1999; Osaki et al. 2000; Eurvilaichit tunity to maximize other more definitive treatments of
et al. 2000). The management and long-term follow- their lung cancer. With newer tools and techniques we
up of bronchial artery embolization is limited by the can provide much more advanced care to patients
few cases of lung cancer managed in almost all today than ever before. Understanding what can be
studies. Much of the information used must be done is the responsibility of treating physicians.
extrapolated to the lung cancer population. Consultation with a team of experts at your facility or
Surgery would appear to be the most definitive a regional referral center will allow the quickest
therapeutic modality available. Retrospective studies assessment of a patient’s complaints and the most
demonstrate good long-term results with surgical rapid institution of therapeutic measures.
resection of the source of bleeding (Knott-Craig et al.
1993; Bobrowitz et al. 1983). This route should be
cautioned in that limited information regarding sur-
References
gical resection of a bleeding source as lung cancer is
available. If a lung cancer was previously diagnosed,
Agusti AGN, Cardus J, Roca J, Grau J, Xauber A, Rodriguez
surgical resection should have been considered had Roisin R (1997) Ventilation–perfusion mismatch in patients
the patient been a surgical candidate and the tumor with pleural effusion. Effects of thoracentesis. Am J Rep
amenable to surgical resection. If a tumor was Crit Care Med 156:1205–1209
590 M. J. Simoff

Alexiou C, Neuhaus H, Kau RJ, Hauck R, Classen M (1998) Colt HG, Dumon J-F (1991) Airway obstruction in cancer: the pros
Treatment of esophagorespiratory fistula by insertion of an and cons of stents. J Respir Dis 12:741–744, 746, 748–749
esophageal Montgomery and tracheal dynamic stent after Colt HG, Meric B, Dumon JF (1992) Double stents for
failure of conventional endoprosthesis. J Otorhinolaryngol carcinoma of the esophagus invading the tracheo-bronchial
Relat Spec 60:51–54 tree. Gastrointest Endosc 38:485–489
Arabian A, Spagnolo SV (1984) Laser therapy in patients with Conlan AA, Hurwitz SS (1983) Management of massive
primary lung cancer. Chest 86:519–523 hemoptysis with the rigid bronchoscope and cold saline
ATS guidelines: management of malignant pleural effusions lavage. Thorax 35:901–904
(2000) Am J Respir Crit Care Med 162:1987 Cook TA, Dehn TC (1996) Use of covered expandable metal
Beamis JF Jr, Vergos K, Rebeiz EE (1991) Endoscopic laser stents in the treatment of oesophageal carcinoma and
therapy for obstructing tracheobronchial lesions. Ann Otol tracheo-oesophageal fistula. British J Surg 83:1417–1418
Rhinol Laryngol 100:413–419 Cooper JD, Pearson FG, Patterson GA (1989) Use of silicone
Bense L (1990) Intrabronchial selective coagulative treatment stents in the management of airway problems. Ann Thorac
of hemoptysis. Report of three cases. Chest 97(4):990–996 Surg 47:371–378
Bobrowitz ID, Ramakrishna S, Shim YS (1983) Comparison of Corey R, Hla KM (1987) Major and massive hemoptysis:
medical v surgical treatment of major hemoptysis. Arch reassessment of conservative management. Am J med Sci
Intern Med 143:1343–1346 294:301–309
Bolliger CT, Probst R, Tschopp K (1993) Silicone stents in the Cortese DA, Edell ES (1993) Role of phototherapy, laser
management of inoperable tracheobronchial stenoses: therapy, brachytherapy, and prosthetic stents in the manage-
indications and limitations. Chest 104:1653–1659 ment of lung cancer. Clin Chest Med 14:149–159
Boutin C, Viallat JR, Cargnino P, Farisse P (1981) Thoracos- Credle WF Jr, Smiddy JF, Elliott RC (1974) Complications of
copy in malignant pleural effusions. Am Rev Respir Dis fiberoptic bronchoscopy. AM Rev Resp Dis 109:67–72
124:588 Desai SJ, Mehta AC, Vanderbug Medendorp S (1988) Survival
Brown NE, Zamel N, Aberman A (1978) Changesin pulmonary experience following Nd:YAG laser photoresection for
mechanics, gas exchange following thoracentesis. Chest primary bronchogenic carcinoma. Chest 94:939–944
74:540 Diaz-Jimenez JP, Munoz EF, Ballarin JIM (1994) Silicone
Brutinel WM, Cortese DA, McDougall JC (1987) A two-year stents in the management of obstructive tracheobronchial
experience with the neodymium-YAG laser in endobron- lesions: 2-year experience. J Bronchol 1:15–18
chial obstruction. Chest 91:159–165 Doona M, Walsh D (1998) Benzonate for opioid-resistant
Cahill BC, Ingbar DH (1994) Massive hemoptysis. Assessment cough in advanced cancer. Palliat Med 12:55–58
and management. Clin Chest Medicine 15:147–167 Dumon JF (1990) A dedicated tracheobronchial stent. Chest
Cancer Facts and Figures (2001) American Cancer Society. 97:328–332
Atlanta, Ga Dumon JF, Reboud E, Garbe L (1982) Treatment of
Canto A, Blasco E, Casillas M (1977) Thoracoscopy in the tracheobronchial lesions by laser photoresection. Chest
diagnosis of pleural effusion. Thorax 32:550 81:278–284
Cavaliere S, Foccoli P, Farina PL (1988) Nd:YAG laser Eichenhorn MS, Kvale PA, Miks VM (1986) Initial combina-
bronchoscopy. Chest 94:15–21 tion therapy with YAG laser photoresection and irradiation
Cavaliere S, Foccoli P, Toninelli C (1994) Nd:YAG laser for inoperable non-small cell carcinoma of the lung: a
therapy in lung cancer: an 11-year experience with 2, 253 preliminary report. Chest 89:782–785
applications in 1, 585 patients. J Bronchol 1:105–111 Emerson GL, Emerson MS, Sherwood CE (1959) The natural
Chawla M, Getzen T, Simoff M (2009) Medical pneumonect- history of carcinoma of the lung. J Thorac Surg 37:291
omy: interventional bronchoscopic and endovascular man- Escalante CP, Martin CG, Elting LS (1996) Dyspnea in cancer
agement of massive hemoptysis due to pulmonary artery patients. Etiology, resource utilization, and survival impli-
pseudoaneurysm, a consequence of endobronchial bra- cations in a managed care world. Cancer 78:1314–1319
chytherapy. Chest 135:1355–1358 Escudero Bueno C, Garcia Clemente M, Cuesta Castro B
Chen YM, Shih JF, Yang KY, Lee YC, Perng RP (2000) (1990) Cytologic, bacteriologic analysis of fluid, pleural
Usefulness of pigtail catheter for palliative drainage of biopsy specimens with Cope’s needle. Study of 414
malignant pleural effusions in cancer patients. Support Care patients. Arch Intern Med 150:1190
Cancer 8:423–426 Estenne M, Yernault JC, De Troyer A (1983) Mechanism of
Chernow B, Sahn SA (1977) Carcinomatous involvement of the relief of dyspnea after thoracocentesis in patients with large
pleura: an analysis of 96 patients. Am J Med 63:695 pleural effusions. Am J Med 74:813–819
Chute CG, Greenberg ER, Baron J (1985) Presenting conditions Eurvilaichit C, Supasinsathit T, Saenghirunvattana S (2000)
of 1539 population-based lung cancer patients by cell type, Bronchial artery embolization for hemoptysis. J Med Assoc
stage in New Hampshire, Vermont. Cancer 56:2107 Thai 83:590–600
Clarke CP, Ball DL, Sephton R (1994) Follow-up of patient Fentiman IS, Rubens RD, Hayward JL (1983) Control of
having Nd:YAG laser resection of bronchostenotic lesions. pleural effusions in patients with breast cancer. A random-
J Bronchol 1:19–22 ized trial. Cancer 52:737
Cohen S, Hossain S (1966) Primary carcinoma of the lung: a Fentiman IS, Rubens RD, Hayward JLA (1986) Comparison of
review of 417 histologically proved cases. Dis Chest 49:626 intracavitary talc, tetracycline for the control of pleural
Colt HG, Dumon JF (1993) Tracheobronchial stents: indica- effusions secondary to breast cancer. Eur J Cancer Clin
tions and applications. Lung Cancer 9:301–306 Oncol 22:1079
Advances in Supportive and Palliative Care for Lung Cancer Patients 591

Freitag L, Eicker K, Donovan TJ (1995) Mechanical properties Ikeda S (1995) Flexible bronchofiberscope. Ann Otol Rhinol
of airway stents. J Bronchol 2:270–278 Respir 62:148–150
Freitag L, Tekolf E, Steveling H, Donovan TJ, Stamatis G (1996) Ikeda S (1970) Flexible bronchofiberscope. Ann Otol Rhinol
Management of malignant esophago-tracheal fistulas with Laryngol 79:916–923
airway stenting and double stenting. Chest 110:1155–1160 Jacox A, Carr DB, Payne R (1994) Management of cancer pain:
Frost JK, Ball WC Jr, Levin MI (1984) Early lung cancer clinical practice guidelines No. 9. Rockville, MD: Agency
detections: results of the initial (prevalence) radiologic and for Health Care Policy and Research, U.S. Department of
cytologic screening in the Johns Hopkins study. Am Rev Health and Human Services, Public Health Service AHCPR
Respir Dis 130(4):549 Publication No. 94-0592
Furuse K, Fukuoka M, Kato H (1993) A prospective phase II Jain PR, Dedhia HV, Lapp NL, Thompson AB, Frich JC Jr
study on photodynamic therapy with Photofrin II for (1985) Nd:YAG laser followed by radiation for treatment of
centrally located early-stage lung cancer: the Japan lung malignant airway lesions. Lasers Surg Med 5:47–53
cancer photodynamic therapy study group. J Clin Oncol Jean-Baptiste E (2000) Clinical assessment, management of
11:1852–1857 massive hemoptysis. Crit Care Med 28:1642
Gaer JA, Tsang V, Khaghani A (1992) Use of endotracheal Johnston WW (1985) The malignant pleural effusion: a review
silicone stents for relief of tracheobronchial obstruction. of cytopathological diagnoses of 584 specimens from 472
Ann Thorac Surg 54:512–516 consecutive patients. Cancer 56:905
Galbraith S, Fagan P, Perkins P, Lynch A, Booth S (2010) Does Karetzky MS, Kothari GA, Fourre JA, Khan AU (1978) Effect
the use of a handheld fan improve chronic dyspnea? A of thoracentesis on arterial oxygen tension. Respiration
randomized, controlled crossover trial. J Pain Symptom 36:96
Manag 29:831–838 Kennedy L, Rusch VW, Strange C (1994) Pleurodesis using
Gelb AF, Zamel N, Colchen A (1992) Physiologic studies of talc slurry. Chest 106:342
tracheobronchial stents in airway obstruction. Am Rev Knott-Craig CJ, Oostuizen JG, Rossouw G, Joubert JR, Barnard
Respir Dis 146:1088–1090 PM (1993) Management and prognosis of massive hemopt-
Gerasin VA, Shafirovsky BB (1988) Endobronchial electro- ysis. Recent experience with 120 patients. J Thorc Cardio-
surgery. Chest 93:270–274 vasc Surg 105:394–397
Gong H Jr, Salvatierra C (1981) Clinical Efficacy of early, Koeda K, Ishida K, Sato N, Ikeda K, Kimura Y, Saito K (1997)
delayed fiberoptic bronchoscopy inpatients with hemopty- Nippon kyobu geka gakkai zasshi. J Jpn Assoc Thoracic
sis. Am Rev Respir Dis 124:221 Surg 45:1169–1172
Grippi MA (1990) Clinical aspects of lung cancer. Semin Krell WS, Rodarte JR (1985) Effects of acute pleural effusion
Roentgenol 25(1):12 on respiratory system mechanics in dogs. J Appl Physiol
Hamed H, Fentiman IS, Chaudary MA, Rubens DS (1989) 59:1458
Comparison of intracavitary bleomycin, talc for the control Kvale PA, Eichenhorn MS, Radke JR (1985) YAG laser
of pleural effusions secondary to carcinoma of the breast. Br photoresection of lesions obstructing the central airways.
J Surg 76:1266 Chest 87:283–288
Hartman DL, Gaither JM, Kesler KA (1993) Comparison of Kvale P, Simoff M, Prakash U (2003) Palliative care: diagnosis
insufflated talc under thoracoscopic guidance with standard and management of lung cancer: evidence-based guidelines.
tetracycline, bleomycin pleurodesis for control of malignant Chest 123:284S–311S
pleural effusions. J Thorac Cardiovasc Surg 105:743 Lam S (1994) Photodynamic therapy of lung cancer. Semin
Hayata Y, Kato H, Konaka C (1993) Photodynamic therapy Oncol 21:15–19
(PDT) in early stage lung cancer. Lung Cancer 9:287–294 Lamy P, Canet B, Martinet Y, Lamaze R (1980) Evaluation of
Heffner JE, Standerfer RJ, Torstveit J, Unruh L (1994) Clinical diagnostic means in pleural effusions (from two hundred
efficacy of doxycycline for pleurodesis. Chest 105:1743 observations) (author’s transl). Poumon Coeur 36:83
Hetzel MR, Smith SG (1991) Endoscopic palliation of Lan RS, Lo SK, Chuang ML (1997) Elastance of the pleural
tracheobronchial malignancies. Thorax 46:325–333 space: a predictor for the outcome of pleurodesis in patients
Hetzel MR, Millard FJ, Ayesh R (1983) Laser treatment for with malignant pleural effusion. Ann Intern Med 126:768
carcinoma of the bronchus. Br Med J 286:12–16 Le Roux BT (1968) Bronchial carcinoma. ES Livingstone,
Hirsch A, Ruffie P, Nebut M (1979) Pleural effusion: laboratory Edinburgh, p 127
tests in 300 cases. Thorax 34:106 Light RW, Jenkinson SG, Minh V, George RB (1980)
Hirschberg B, Biran I, Glazer M, Kramer MR (1997) Hemopt- Observations on pleural pressures as fluid is withdrawn
ysis: etiology, evaluation, outcome in a tertiary referral during thoracentesis. Am Rev Respir Dis 121:799
hospital. Chest 112:440 Light RW, Stansbury DW, Brown SE (1986) The relationship
Hoegler D (1997) Radiotherapy for palliation of symptoms in between pleural pressures, changes in pulmonary function
incurable cancer. Curr Probl Cancer 21:129–183 after therapeutic thoracentesis. Am Rev Resp Dis 133:658
Hoegter D (1997) Radiotherapy for palliation of symptoms in Livingston RB, McCracken JD, Trauth CJ, Chen T (1982)
incurable cancer. Curr Probl Cancer 21(3):129 Isolated pleural effusion in small cell lung carcinoma:
Hsu C (1987) Cytologic detection of malignancy in pleural favorable prognosis. Chest 81:208
effusion: a review of 5, 255 samples from 3, 811 patients. Loddenkemper R (1981) Thoracoscopy: results in non-cancer-
Diagn Cytopathol 3:8 ous, idiopathic pleural effusions. Poumon Coeur 37:261
Hyde L, Hyde CI (1974) Clinical manifestations of lung cancer. Loddenkemper R (1998) Thoracoscopy state of the art. Eur
Chest 65:299 Respir J 11:213
592 M. J. Simoff

Loddenkemper R, Grosser H, Gabler A (1983) Prospective Osaki S, Nakanishi Y, Wataya H (2000) Prognosis of bronchial
evaluation of biopsy methods in the diagnosis of malignant artery embolization in the management of hemoptysis.
pleural effusions: intrapatient comparison between pleural Respiration 67:412
fluid cytology, blind needle biopsy and thoracoscopy. Am Patz EF, McAdams HP, Erasmus JJ (1998) Sclerotherapy for
Rev Respir Dis 127(suppl 4):114 malignant pleural effusions: a prospective randomized trial
Luomanen RKJ, Watson WL (1968) Autopsy findings. In: of bleomycin vs doxycycline with small-bore catheter
Watson WL (ed) Lung cancer: a study of five thousand drainage. Chest 113:1305
memorial hospital cases. St Louis, MO: CV Mosby Co, Petrou M, Kaplan D, Goldstraw P (1995) The management of
pp 504–510 recurrent malignant pleural effusions: the complementary
Macha HN, Becker KO, Kemmer HP (1994) Pattern of failure role of talc pleurodesis, pleuro-peritoneal shunting. Cancer
and survival in endobronchial laser resection: a matched 75:801
pair study. Chest 105:1668–1672 Petrovich Z, Stanley K, Cox JD (1981) Radiotherapy in the
Magee G, Williams MH Jr (1982) Treatment of massive management of locally advanced lung cancer of all cell
hemoptysis with intravenous Pitressin. Lung 160:165–169 types: final report of randomized trial. Cancer 48:
Maiwand MO, Homasson JP (1995) Cryotherapy for tracheo- 1335–1340
bronchial disorders. Clin Chest Med 16:427–443 Pien GW, Gant MJ, Washam CL, Sterman DH (2001) Use of an
Mal H, Rullon I, Mellot F (1999) Immediate, long-term results implantable pleural catheter for trapped lung syndrome in
of bronchial artery embolization for life-threatening hem- patients with malignant pleural effusion. Chest 119:1641–1646
optysis. Chest 115:996 Poe RH, Israel RH, Utell MJ (1984) Sensitivity, specificity,
Martensson G, Pettersson K, Thiringer G (1985) Differentiation predictive values of closed pleural biopsy. Arch Intern Med
between malignant, non-malignant pleural effusion. Eur J 144:325
Respir Dis 67:326 Pollak JS, Burdge CM, Rosenblatt M, Houston JP, Hwu WJ,
Martinez-Moragon E, Aparicio J, Rogado MC (1997) Pleurod- Murren J (2001) Treatmet of malignant pleural effusion
esis in malignant pleural effusions: a randomized study of with tunneled long-term drainage catheters. J Vasc Interv
tetracycline versus bleomycin. Eur Respir J 10:2380 Radiol 12:201–208
Mathisen DJ, Grillo HC (1989) Endoscopic relief of malignant Ponn RB, Blancaflor J, D’Agostino RS, Kiernan ME, Toole
airway obstruction. Ann Thorac Surg 48:469–475. A five- AL, Stern H (1991) Pleuroperitoneal shunting for intracta-
year experience with 1,396 applications in 1,000 patients ble pleural effusions. Ann Thor Surg 51:605–609
McDougall JC, Corese DA (1983) Neodymium-YAG laser Pope AR, Joseph JH (1989) Pleuroperitoneal shunt for pneumo-
therapy of malignant airway obstruction: a preliminary nectomy cavity malignant effusion. Chest 96:686–688
report. Mayo Clin Proc 58:35–39 Prakash UBS, Reiman HM (1985) Comparison of needle
McGuiness G, Beacher JR, Harkin TJ (1994) Hemoptysis: biopsy with cytologic analysis for the evaluation of pleural
prospective high-resolution CT/bronchoscopic correlation. effusions: analysis of 414 cases. Mayo Clin Proc 60:158
Chest 105:1155 Prakash UB, Stubbs SE (1991) The bronchoscopy survey: some
Mehta AC (2008) AERO self expanding hybrid stent for airway reflections. Chest 100:1660–1667
stenosis. Expert Rev Med Devices 5:553–557 Pulsiripunya C, Youngchaiyud P, Pushpakom R (1996) The
Mehta AC, Golish JA, Ahmad M (1985) Palliative treatment of efficacy of doxycycline as a pleural sclerosing agent in
malignant airway obstruction by Nd-YAG laser. Cleve Clin malignant pleural effusion: a prospective study. Respirology
Q 52:513–524 1:69
Menzies R, Charbonneau M (1991) Thoacoscopy for the Putnam JB, Light RW, Rodriquez RM (1999) Randomized
diagnosis of pleural disease. Ann Intern Med 114:271 comparison of indwelling pleural catheter with doxycycline
Milanez Campos JR, Werebe EC, Vargas FS (1997) Respira- pleurodesis in the management of malignant pleural effu-
tory failure due to talc. Lancet 349:251–252 sion. Cancer 86:1992–1999
Miller RR, McGregor DH (1980) Hemorrhage from carcinoma Rehse DH, Aye RW, Florence MG (1996) Respiratory failure
of the lung. Cancer 46:200 following talc pleurodesis. Am J Surg 162:2023–2026
Moffett MJ, Ruckdeschel JC (1992) Bleomycin, tetracycline in Reich H, Beattie EJ, Harvey JC (1993) Pleuroperitoneal shunt
malignant pleural effusions: a review. Semin Oncol 19:59 for malignant pleural effusions: a one-year experience.
Moghissi K, Dixon K, Stringer M (1999) The place of Semin Surg Onc 9:160–162
bronchoscopic photodynamic therapy in advanced unresec- Rodriguez-Panadero F, Lopez-Mejias L (1989) Low glucose,
table lung cancer: experience of 100 cases. Eur J Cardioth- pH levels in malignant effusions; diagnostic significance,
roac Surg 15:1–6 prognostic value in respect to pleurodesis. Am Rev Respir
Montgomery WW (1965) T-tube tracheal stent. Arch Otolar- Dis 139:663
yngol 82:320–321 Ross DJ, Mohsenifar Z, Koerner SK (1990) Survival charac-
Morice RC, Ece T, Ece F, Keus L (2001) Endobronchial argon teristics after Neodymium:YAG laser photoresection in
plasma coagulation for treatment of hemoptysis and advanced stage lung cancer. Chest 98:581–585
neoplastic airway obstruction. Chest 119:781–787 Sahebjami H (1976) Iced saline lavage during bronchoscopy.
Muller NL (1994) Hemoptysis: High-resolution CT vs bron- Chest 69:131
choscopy. Chest 105:982 Schlehe H, Fritsche HM, Daum S (1984) A new method for
Oldenburg FA Jr, Newhouse MT (1979) Thoracoscopy: a safe treatment of bronchial and lung bleeding. In: Nakhosteen JA,
accurate diagnostic procedure using the rigid thoracoscope, Maassen W (eds) Bronchology: Research, Diagnostic, and
local anesthesia. Chest 75:45 Therapeutic Aspects. Springer 1981, Nijhoff, Haag, p 111
Advances in Supportive and Palliative Care for Lung Cancer Patients 593

Schulze M, Boehle AS, Kurdow R, Dohrmann P, Henne-Bruns Toty L, Personne C, Colchen A (1981) Bronchoscopic
D (2001) Effective treatment of malignant pleural effusion management of tracheal lesions using the neodymium
by minimal invasive thoracic surgery: thoracoscopic talc yttrium aluminum garnet laser. Thorax 36:175–178
pleurodesis and pleuroperitoneal shunts in 101 patients. Ann Tsukamoto T, Sasaki H, Nakamura H (1989) Treatment of
Thor Surg 71:1809–1812 hemoptysis patients by throbin and fibrinogen–thrombin
Set PAK, Flower CDR, Smith IE (1993) Hemoptysis: compar- infusion therapy using a fiberoptic bronchoscope. Chest
ative study of the role of CT, fiberoptic bronchoscopy. 96:473–476
Radiology 189:677 van de Molengraft FJ, Vooijs GP (1988) The interval between
Sonett JR, Keenan RJ, Ferson PF (1995) Endobronchial the diagnosis of malignancy, the development of effusions,
management of benign, malignant, and lung transplantation with reference to the role of cytologic diagnosis. Acta Cytol
airway stenosis. Ann Thorac Surg 59:1417–1422 32:183
Spivak F, Katariya K, Lo AY, Harvey JC (1996) Malignant Walker-Renard P, Vaughan LM, Sahn SA (1994) Chemical
tracheo-esophageal fistula: use of esophageal endoprosth- pleurodesis for malignant pleural effusions. Ann Intern Med
esis. J Surg Oncol 63:65–70 120:56
Stanopoulos IT, Beamis JF Jr, Martinez FJ (1993) Laser White RI Jr (1999) Bronchial artery embolotherapy for control
bronchoscopy in respiratory failure from malignant airway of acute hemoptysis: analysis of outcome. Chest 115:912
obstruction. Crit Care Med 21:386–391 World Health Organization. Cancer pain relief and palliative
Starr RL, Sherman ME (1991) The value of multiple prepara- care: Report of a WHO expert committee. World Health
tions in the diagnosis of malignant pleural effusions. Organization Technical Report Series, 804. 1990, 1–75
A cost–benefit analysis. Acta Cytol 35:533 World Health Organization, Geneva, Switzerland
Storey DD, Dines DE, Coles DT (1976) Pleural effusion. Worth H, Breuer HWM, Charchut S, Trampisch HR, Glaenzer K
A diagnostic dilemma. JAMA 236:2183 (1987) Endobronchial versus intravenous application of
Strange C (1991) Double-lumen endotracheal tubes. Clin Chest glypressin for the therapy and prevention of lung bleeding
Med 12:497 during bronchoscopy. Am Rev Resp Dis 135(4 part 2):A-108
Sutedja T, Lam S, LeRiche JC (1994) Response and pattern of Yin AC, Chan AT, Lee TW (1996) Thoracoscopic talc
failure after photodynamic therapy for intraluminal stage I insufflation versus talc slurry for symptomatic malignant
lung cancer. J Bronchol 1:295–298 pleural effusion. Ann Thorac Surg 62:1655
Sutedja G, Schramel F, van Kralingen K (1995) Stent Zavala DC (1976) Pulmonary hemorrhage in fiberoptic trans-
placement is justifiable in end-stage patients with malignant bronchial biopsy. Chest 70:584–588
airway tumours. Respiration 62(3):148–150 Zimmer PW, Hill M, Casey K (1997) Prospective randomized
Todd TRJ, Delarue NC, Ilves R (1980) Talc poudrage for trial of talc slurry vs bleomycin in pleurodesis for
malignant pleural effusion. Chest 78:542 symptomatic malignant pleural effusions. Chest 112:430
Hematological Toxicity in Lung Cancer
Francesc Casas, Ferran Ferrer, and Núria Viñolas

Contents Abstract
The toxicity of tumor cells after chemo and
1 Introduction.............................................................. 598 radiotherapy, administered either alone or in
2 Toxicity in Chemotherapy ...................................... 598 combination is dose-dependent. Aggression to the
bone marrow, which is expressed by a reduction in
3 Toxicity in Radiotherapy ........................................ 600
circulating blood cells, is often the main dose-
4 Hematologic Toxicity After Combined Chemo limiting toxicity because of the risks of anemia,
and Radiotherapy .................................................... 600
bleeding, and infection. Prophylactic treatment
5 Preventive or Support Treatment of Hematologic with granulocyte-colony stimulating factors
Toxicity in Lung Cancer......................................... 603 (G-CSFs) is available to reduce the risk of
References.......................................................................... 606 chemotherapy-induced neutropenia. In 2005, a
European Guidelines Working Party was set up
by the European Organization for Research and
Treatment of Cancer (EORTC) to systematically
review the data published on the appropriate use of
G-CSF in adult patients receiving chemotherapy.
An update of this review was published in 2010.
The ASCO has made evidence level II recommen-
dations concerning the treatment of anemia with
r-Hu-EPO. For patients with chemotherapy-
associated anemia the Committee continues to
recommend the implementation of an erythropoi-
esis-stimulating agent (ESA) when hemoglobin
F. Casas (&) (Hb) values approach or fall below 10 g/dL, to
Radiation Oncology Department,
Hospital Clínic i Universitari, increase Hb values and decrease transfusions. An
Villarroel 170, 08036 Barcelona, Spain individual patient data-analysis has shown that
e-mail: fcasas@clinic.ub.es ESA increase the mortality in all patients with
F. Ferrer cancer, and a similar increase might exist in
Radiation Oncology Department, patients on chemotherapy. Finally, only a few trials
Institut Català d’Oncologia, have examined indications for ESA similar to the
Gran Via de les Corts Catalanes,
Bellvitge, Spain indications approved by the Food and Drug
Administration.
N. Viñolas
Medical Oncology Department,
Hospital Clínic i Universitari,
Villarroel 170, 08036 Barcelona, Spain

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 597
DOI: 10.1007/174_2011_271, Ó Springer-Verlag Berlin Heidelberg 2011
598 F. Casas et al.

cells are also found in the spleen and circulate in the


1 Introduction blood.
Bone marrow dysfunction in neoplastic processes
This chapter will describe the normal physiology of may be due to different etiologies:
bone marrow followed by a synthesis of the current 1. Depletion or direct lesions of the hematopoietic
knowledge of the toxicity of these two treatments stem cells.
either alone or in combination. Lastly, support treat- 2. Functional or structural damage of the stroma or
ments and the management of these secondary effects the microcirculation.
are proposed. 3. Lesion of other collaborator cells which have a
The toxicity of tumor cells after chemo and regulator function or hemostasis.
radiotherapy, administered either alone or in combi- The consequences of the aggression of cytotoxic
nation is dose-dependent. Aggression to the bone and radiotherapeutic treatment to the bone marrow
marrow, which is expressed by a reduction in circu- should, therefore, be understood within the context of
lating blood cells, is often the main dose-limiting the previously described mechanisms. Nonetheless, it
toxicity because of the risks of anemia, bleeding, and may be difficult to elucidate the most important
infection. Strategies aimed at protecting the hemato- variables due to limitations in the evaluation of both
poietic cells or the stroma of the bone marrow from the structure and the bone marrow function. Periph-
death induced by the treatment, and the acceleration eral determination of the blood cells fails to demon-
of hematopoiesis after treatment, may theoretically strate the true extension of bone marrow suppression
allow more intensive treatments in lung cancer (LC) or its capacity to tolerate additional cytotoxic therapy
without the above-mentioned associated risks. It is mainly because of the capacity of the bone marrow to
necessary to know the structure and function of the transitorily compensate the aggression. To evaluate
bone marrow as an organ to know the true impact of several quantitative and functional aspects of the bone
individual or combined, sequential or concurrent marrow cultures of progenitor cells, histopathologic
treatment and thereby act accordingly. Thus, the studies (bone marrow aspirate and biopsy), and
pluripotent stem cells replicate and differentiate in determined radioisotopes or stromal cell cultures may
lymphoid or myeloid lines through a complex process be used, albeit to a limited extent.
regulated by a network of hematopoietic growth
factors as well as by cellular interactions. The cascade
through myeloid differentiation leads to the erythro- 2 Toxicity in Chemotherapy
cytes, platelets, granulocytes, and macrophages, while
lymphoid differentiation leads to T and B cells. The myelosuppression directly caused by chemo-
Families of growth factors (or cytokines) which therapy (CH) not only depends on the agent used but
control these processes of replication and differenti- also on patient-dependent factors, such as age and
ation have been identified. The hematopoietic pro- general status. Important factors in relation to the type
genitor cells and their daughter cells are enveloped in of CH administered are the dosis, the interval of the
a stroma of endothelial cells, adventitial cells, fibro- dosis, the route of administration, or the use of a
blasts, macrophages, and fat cells in the sinus of the single or several antitumoral agents. On the other
bone marrow. This microscopic medium physically hand, the site of action of the antineoplastic drug
supports and directs the development of the replica- within the cellular cycle also appears to influence
tion process. In addition, the geographic distribution myelosuppression.
of the bone marrow is particularly relevant to know Damage results from a depletion in the total
the possible local effects of radiotherapy in the number of stem cells (the stem cell pool) with a late
treatment of LC. The most functional and important myelosuppression pattern which takes place when the
localizations are the pelvis, the vertebrae (these two peripheral blood cells die and cannot be replaced.
represent 60% of the total bone marrow), as well as That is to say that myelotoxicity by CH agents pro-
the ribs, the sternum, the cranium, the scapula, and the duces a decrease in the production of blood cells more
proximal portions of the femur and humeral bones. than an immediate elimination of the peripheral cells
It should be remembered that hematopoietic stem (Ratain et al. 1990).
Hematological Toxicity in Lung Cancer 599

Because of differences in the peripheral blood half Patient-related risk factors should be evaluated in
life, drugs that induce myelosuppression first result in the overall assessment of FN risk before administer-
leukopenia followed by thrombocytopenia with the ing each cycle of CH. Particular consideration should
first former generally being more severe than the be given to the elevated risk of FN for elderly patients
latter. Thus, the nadir for neutrophils and platelets is (aged 65 and over). Other adverse risk factors that
normally between 7 and 15 days after drug adminis- may influence FN risk include advanced stage of
tration. For most of the compounds, neutropenia and disease, experience of previous episode(s) of FN, lack
thrombocytopenia are reversible and not accumula- of G-CSF use, and absence of antibiotic prophylaxis.
tive. In addition to the direct cytotoxicity at the level The risk of FN associated with CH regimens must be
of the progenitor cells, at an erythrocytic level taken into account when evaluating the need for
implicating blood cells with a more prolonged half prophylactic intervention. The new CH regimens with
life, the mechanism involved may be direct hemolysis the addition of targeted agents have been shown to
of the red blood cells after the administration or a improve survival. This is the case of the addition of
decrease in the production of endogenous erythro- cetuximab or bevacizumab to chemotherapy in
poietin due to chronic renal insufficiency by cisplatin NSCLC patients (Pirker et al. 2009; Reck et al. 2009).
(Pivot et al. 2000). The pluripotent stem cells are An increased incidence of FN has been reported in
protected from the toxic effects of CH because of their patients receiving bevacizumab and chemotherapy
slow proliferation. compared with CH alone (Sandler et al. 2006). Con-
CH-induced febrile neutropenia (FN) is a poten- sideration should be given to the elevated risk of FN
tially fatal complication of cancer treatment when it when using certain CH regimens such as docetaxel/
heralds infection and sepsis and is most often seen carboplatin (Milward et al. 2003).
during the initial cycles of myelosuppressive therapy One of the main factors of toxicity for a given
(Timmer-Bonte et al. 2005). FN is defined as an CH agent is the pharmacodynamic interaction
absolute neutrophil count (ANC) of \0.5 9 109/L or between the drug and the combination of other an-
\1.0 9 109/L predicted to fall below 0.5 9 109/L ticancerous drugs. Thus, one of the general princi-
within 48 h, with fever or clinical signs of sepsis ples for combining different drugs is that they should
(Crawford et al. 2010). The European Society for have a different limiting toxicity, although a sum of
Medical Oncology (ESMO) defines fever in this set- these effects is normally produced in relation to
ting as a rise in axillary temperature to [38.5°C myelotoxicity. There is, however, an exception to
sustained for at least 1 h. It is suggested that therapy this rule in the case of the combination of paclit-
be initiated if a temperature of[38°C is presented for axel–carboplatin: paclitaxel decreases the platelet
at least 1 h or a reading of [38.5°C is obtained on a toxicity of carboplatin in relation to a non pharma-
single occasion. cokinetic mechanism (Calvert et al. 1999).
Some of the adverse consequences of CH-induced The most frequent schedules of CH currently used
FN may lead to treatment delay and dose reductions in NSCLC include combinations of cisplatin or car-
which may potentially adversely affect tumor control boplatin with some of the new drugs (gemcitabine,
(Khan et al. 2008). For instance, poor outcome in vinorelbine, paclitaxel, docetaxel). All have shown to
cancer patients has been attributed to failure to deliver be similar in regard to efficacy in stage IV, although
planned CH regimens for LC (Radosavljevic et al. the toxicities observed, including hematologic toxic-
2009). ity, differ (Schiller et al. 2002). These combinations
Recognizing patients at risk for complications of of CH cause grades 3 and 4 neutropenia which varies
FN can be achieved using risk indices such as those from 40 to 70% with febrile neutropenia in less than
developed by the Multinational Association for Sup- 10%. Grades 3 and 4 platelet toxicity was observed in
portive Care in Cancer (MASCC) (De Souza et al. 1 to 55% of the patients, with a schedule combining
2008). Using the MASCC score, patients with a score cisplatin and gemcitabine showing a greater percent-
of 21 or more points are considered at high risk of age of thrombocytopenia (Cardenal et al. 1999).
infectious complications. Identifying patients at risk Patients with carboplatin-based chemotherapy are
of bacteremia facilitates appropriate initiation of more likely to experience thrombocytopenia
antibiotics (Klastersky et al. 2010). (Luo et al. 2010).
600 F. Casas et al.

In regard to anemia, the percentages vary from 10 exclusive irradiation using standard fractionation
to 30%, with the schedules based on cisplatin and leads to subclinical, albeit quantifiable, hematologic
gemcitabin or vinorelbin being those producing the toxicity which will be described in greater depth later
greatest percentage of patients with anemia (Kelly when we discuss combined treatment (chemo- and
et al. 2001). radiotherapy) and compare the resulting myelotoxic-
The sequence of administration is also very ity using references from randomized studies related
important since an increase in myelotoxicity has been to radiotherapy alone.
observed when cisplatin is administered before pac-
litaxel. Platelet toxicity is not of note in schemes
including paclitaxel combined with carboplatin sug-
gesting that paclitaxel is protective. The combination 4 Hematologic Toxicity After
of cisplatin and etoposide produces less neutropenia Combined Chemo
than the CAV and CAE schemes, albeit with more and Radiotherapy
anemia (Fukuoka et al. 1991). The profile of hema-
tologic toxicity with the combination of etoposide and The selective action of CH agents for different pop-
carboplatin is similar to that found with the schedule ulations of hematopoietic cells determines the tem-
of cisplatin except with a higher percentage of porary consequences of the tolerance of the bone
thrombocytopenia. marrow to radiation after CH. In addition, when wide
fields are used before CH, the tolerance expected is
poor. This may be due not only to the suppression or
3 Toxicity in Radiotherapy ablation of determined segments or portions of the
bone marrow, but also to the increase in the sensi-
In the case of irradiation in LC, acute toxicity of the tivity of non exposed zones of the bone marrow
bone marrow depends on the volume irradiated, the which, at that time, are in a period of hyperactivity.
dose of radiation, and its rate. Although the com- This is produced in the case of sequential treatments
pensatory mechanisms are mainly relevant for the further complicating the question when referring to
knowledge of long-term effects, some effects are combined treatments of radio and CH. In the case of
acute. Thus, when volumes limited to the bone mar- SCLC, the study by Abrams et al. (1985) is of note.
row are irradiated, such as, for example, 10 to 15%, These authors randomized 42 patients to receive
the remaining bone marrow responds by increasing either CH alone or in combination with thoracic
the population of progenitor cells. This is why the irradiation. In the group receiving combined treat-
bone marrow, as an organ as a whole, is able to ment an increase was observed in both hematopoietic
regenerate the previously irradiated zone in a com- toxicity and in the circulating number of progenitor
pensatory way. This compensatory phenomenon may cells suggesting that the toxicity of concurrent treat-
be observed by factors (CSFs) from the cell stroma ment is additive. It was found that:
suggesting the implication of a humoral mechanism. a. The combination of CH and thoracic radiotherapy
It has been shown that there is an extensive com- produces more hematologic toxicity during the
munication and compensation network in the bone period of irradiation than when CH is administered
marrow after aggression with radiation and this may alone.
be summarized as follows: b. This increase may be explained by a generally
1. Regeneration within the field of irradiation. subclinical, although measurable, toxicity of the
2. Hyperactivity in non irradiated regions. thoracic radiotherapy when administered alone.
3. Extension of the function of bone marrow pro- c. The potential of hematopoietic toxicity by irradi-
duction in previously dormant zones (Tubiana ation itself may vary in relation to the timing, the
et al. 1979). volume of treatment, to the region irradiated, and
This reparation or compensatory capacity of the the treatment fields used. That is, the greater the
bone marrow makes it difficult to clinically observe volume treated and the greater the quantity of
bone marrow toxicity secondary to exclusive radio- the cardiac circuit and bone marrow involved in
therapy treatment in lung cancer. Nonetheless, this the irradiated fields, the greater the toxicity.
Hematological Toxicity in Lung Cancer 601

Table 1 Hematologic toxicity in randomised concurrent hyperfractionated arms on SCLC


Leukopenia Trombocytopenia Anemia
Grade % Grade % Grade %
Jeremic et al. (1995, 1997) 3 21 3 25 3 11
4 11 4 13 4 2
Turrisi et al. (1999) 3 38 3 13 3 23
4 44 4 8 4 5
Takada et al. (2002) 3 51 3 23 3 54
4 38 4 5 4 –

In recent years, the contribution of not only the Concurrent ONce daily vErsus twice daily Radio-
importance of the timing of the administration (early Therapy (CONVERT) study, the Intergroup study
or late) in concurrent combined treatment, but also the (CALGB 30610/RTOG 0538), and the Randomized
alterations of the fractionation (accelerated hyper- trial of chest irradiation in extensive disease small cell
fractionation vs. standard fractionation) in patients lung cancer (REST) study (Favre-Finn et al. 2010)
with SCLC condition changes in hematological tox- which will provide new information related to
icity. Thus, Murray et al. (1993) randomized a group hematologic toxicities secondary to more limited
of patients into two arms of early (in the third week) radiotherapy fields, current technology, and different
versus late (in the 15th week) concurrent irradiation dose fractionation.
and found that although the differences between At the beginning of the 1990s a series of ran-
neutropenia and thrombocytopenia greater than or domized studies in NSCLC were performed evaluat-
equal to grade 3 were not statistically significant for ing both the effectiveness and the toxicity of
either of the treatment arms, they were so in relation concurrent or sequential radioCH versus irradiation
to grade 3 anemia, which was greater in the late alone (Table 2). Firstly, the study by Le Chevalier
administration (P \ 0.03). et al. (1991) was of note. In this study 353 patients
In a study by Jeremic et al. (1997), 107 patients were randomized to receive 65 Gy of irradiation
were randomized to receive either CH plus early hy- alone versus the same irradiation preceded by three
perfractionated radiotherapy (weeks 1 to 4) with cycles of vindesine, lomustine, cisplatin, and cyclo-
concurrent CH versus late administration (weeks 6 to phosphamide. The group receiving irradiation alone
9) and did not find statistically significant differences showed threefold less hematologic toxicity than the
in hematologic toxicity. In the same year the group of group administered combined therapy. Dillman et al.
the EORTC (Gregor et al. 1997) published another (1990) randomized 155 patients to receive two cycles
randomized study in patients with limited stage SCLC of cisplatin and vinblastine followed by 60 Gy of
comparing sequential radioCH versus alternating irradiation versus radiotherapy alone at the same
treatment and reported that the latter schedule was as doses. Although the hematologic toxicity in this study
effective as the sequential administration but caused was not correctly explained, it was of note that neu-
greater grades 3 and 4 hematologic toxicity. tropenic infection was more prevalent in the patients
Turrisi et al. (1999) carried out a randomized study receiving CH with double the number of admissions
comparing concurrent CH with hyperfractionated due to severe infections versus the patients adminis-
radiotherapy versus the same CH with standard frac- tered irradiation alone.
tionated radiotherapy and found greater toxicity in the In a study by Trovo et al. (1992) 173 stage III
treatment with hyperfractionated radiotherapy. Lastly, patients were randomized to receive 45 Gy versus the
Takada et al. (2002) randomized concurrent versus same irradiation administered concurrently with a
sequential chemoradiotherapy and observed greater daily dose of 6 mg/m2 of cisplatin. The hematologic
hematological toxicity in the first treatment arm toxicity of the combined treatment was only slightly
(Table 1). Finally, in SCLC ongoing clinical trials superior to that of radiotherapy alone. In 1993,
addressing radiotherapy-related questions include the Schaake-Koning et al. (1992) randomized 331 patients
602 F. Casas et al.

Table 2 Hematologic toxicity in randomised trials on NSCLC


Hematologic toxic RT group CH ? RT group (monthly CH ? RT group (daily
effect (%) CH) (%) CH) (%)
Le Chevalier et al. (1991) Grade 2–5 1.4 4.2 –
(sequential)
Trovo et al. (1992) (concurrent) Hemoglobin 1.7 – 2.3
(grade 1–2)
Leukopenia 1.1 – 1.7
Schaake-Konig et al. (1992) Leukopenia (grade 3.3 6.6 (weekly CH) 14.5
(concurrent) 3–4)
Trombopenia 0.6 0.9 (weekly CH) 1.8
(grade 3–4)
Dillman et al. (1990) Neutropenia 3 7 –
(sequential)

to receive 56 Gy administered by split-course or the greater than sequential. This was corroborated by Fu-
same radiotherapy plus 30 mg/m2 of cisplatin admin- ruse et al. (1999) in a study in which 320 stage III
istered each week of irradiation versus the same total SCLC patients were randomized to receive concurrent
dosis of irradiation administered continuously with a treatment with cisplatin, vindesine, and mitomycin and
daily dosis of 6 mg/m2 of cisplatin during irradiation. It 56 Gy administered by split-course versus the same
was found that grades 3–4 hematologic toxicity was CH and one continuous dosis of 56 Gy. Greater
fourfold greater in the group with concurrent admin- immunosuppression was also observed in the concur-
istration with weekly cisplatin compared to radio- rent treatment arm.
therapy alone and was double in the concurrent A second similar trial published involved concur-
treatment with daily versus weekly CH. rent versus sequential radioCH with cisplatin and
Sause et al. (1995) published a randomized study vinorelbine in locally advanced NSCLC (Zatloukal
on whether patients receiving CH followed by irra- et al. 2004). Grade 3 or 4 toxicity was more frequent
diation showed longer survival than hyperfractionated in arm A than in arm B, with a significantly greater
radiotherapy or irradiation with standard fractionation incidence of leukopenia (53 vs. 19%, P = 0.009).
in patients with stage III NSCLC. Neutropenic A new combination of treatment has also been
toxicity greater than grade 3 was presented in 50% of investigated with a simultaneous radioCH (paclitaxel
the patients with combined treatment and was null in 60 mg/m2, weekly) compared with radiotherapy alone
the other two treatment arms. Jeremic et al. (1995) after induction CH in inoperable stage IIIA or IIIB
randomized 169 patients to receive hyperfractionated NSCLC: the CTRT99/97 study by the Bronchial
radiotherapy at 1.2 Gy/twice a day up to a total dosis Carcinoma Therapy Group (Huber et al. 2006).
of 64.8 Gy versus the same dosis of irradiation plus Induction CH was well tolerated with 3.8% patients
100 mg of carboplatin on days 1 and 2 and 100 mg of with grade 3 or 4 leukopenia (grade 4, 2.1%).
etoposide on days 1 and 3 of each week of irradiation Hematologic toxicities during radiotherapy alone or
versus a third group in which the same radiotherapy radioCH were both equivalent and without grade
was administered plus 200 mg of carboplatin admin- 3 or 4.
istered on days 1 and 2 and 100 mg of VP-16 on days In a randomized phase II, the Cancer and Leuke-
1 and 5 of the first, third, and fifth week of irradiation. mia Group B (CALGB) studied the effectiveness and
Likewise, the toxicity was greater in the combined tolerance of two cycles of induction CH followed by
treatment, especially in the second group. two additional cycles of the same CH plus concurrent
On demonstration of the greater effectiveness, albeit radiotherapy. The CH used was doublets of cisplatin
with greater hematological toxicity, of sequential with gemcitabine, vinorelbine, and paclitaxel (Vokes
treatment versus exclusive irradiation, the next step et al. 2002) and in this study hematologic toxicity was
was to demonstrate that concurrent administration was presented separately in the induction and also in the
Hematological Toxicity in Lung Cancer 603

Table 3 Hematologic toxicity in randomised trials studying the role of induction CH in concurrent CH-RT on NSCLC
Hematologic toxic Induction RT group CH ? RT
effect group (%) (%) group (%)
Huber et al. (2006) (weekly CH-RT vs. RT alone Leukopenia (grade 3.8 0 0
after induction CH) 3–4)
Anemia (grade – 0 0
3–4)
Vokes et al. (2007) (inmediate CH-RT vs. induction Granulocytopenia Inmediate After
CH-RT) CH-RT induction
Grade 3 18 11 24
Grade 4 20 4 7
Anemia
Grade 3 1 5 12
Grade 4 0 0 0

concurrent treatment. In the first part grade 3–4 further improve survival compared with PE/XRT
granulocytopenia was of note in 50% of the patients alone in patients with stage III inoperable NSCLC. In
in the three treatment arms presented, and in the arm patients receiving docetaxel, 10.9% experienced feb-
with gemcitabine 25% of the patients also presented rile neutropenia (Hanna et al. 2008). A total of 28.8%
grades 3 and 4 thrombocytopenia. In regard to the of patients were hospitalized during docetaxel
toxicity observed with concurrent treatment it was of (vs. 8.1% in the observation arm) and 5.5% died as a
note that notable differences were found in the three result of docetaxel. To date, there is insufficient evi-
treatment arms of the study. Thus, while in the groups dence indicating that treatment extending beyond
treated with gemcitabine and paclitaxel grades 3 and concurrent CH alone further improves survival rates.
4 granulocytopenia were observed in 51 and 53% Many questions remain unanswered in the treatment
respectively, in the group receiving vinorelbine this of stage III stage disease including defining the
hematologic toxicity was seen in 27% of the patients. optimal CH regimen and the utility of lower dose
Platelet toxicity was also found to be greater (50%) in radiosensitizing CH, individualization of the radio-
the group with concurrent treatment with gemcita- therapy dose and fractionation, specifically after sur-
bine. With this background the CALGB made a phase vival improving metaanalysis with accelerated
III trial on immediate concurrent radioCH with car- radiotherapy (Saunders et al. 2010) based on pul-
boplatin area under the concentration–time curve monary function, tumor volumes, and newer radio-
(AUC) of 2 and paclitaxel 50 mg/m2 given weekly therapy technologies.
during 66 Gy of chest radiotherapy, or arm B, which
involved two cycles of carboplatin AUC 6 and plac-
litaxel 200 mg/m2 administered every 21 days fol- 5 Preventive or Support Treatment
lowed by identical radioCH. The accrual was 360 of Hematologic Toxicity in Lung
patients (Vokes et al. 2007). There was no difference Cancer
in survival between the two arms. Adverse events to
treatment during induction CH on arm B included Prophylactic treatment with G-CSF is available to
grade 3 or 4 granulocytopenia in 18 and 20% of reduce the risk of CH-induced neutropenia. However,
patients, respectively. Neutropenia was significantly the use of G-CSF prophylactic treatment varies widely
increased in arm B reflecting the accumulative effect in clinical practice, both in the timing of the therapy
of induction CH (Table 3). and in the patients to whom it is offered. In 2005, a
Finally, a phase III trial by the Hoosier Oncology European Guidelines Working Party was set up by the
Group showed that consolidation docetaxel after European Organization for Research and Treatment of
cisplatin/etoposide and concurrent radiotherapy (PE/ Cancer (EORTC) to systematically review the pub-
XRT) results in increased toxicities but does not lished data available and derive evidence-based
604 F. Casas et al.

recommendations on the appropriate use of G-CSF in Nonetheless, in certain high risk patients with clear
adult patients receiving CH (Aapro et al. 2006). predictive factors of worse outcome (for example in
Nonetheless, with regard to the use of daily G-CSF sepsis, pneumonia, fungal infections, etc.) the use of
versus once-per cycle pegylated G-CSF, additional CSF together with antibiotics may be justified
evidence has emerged since the publication of the last (Bennett et al. 1999). In 2002, a systematic review
EORTC guidelines. In addition, further filgastrim was published on the randomized trials conducted
biosimilar molecules have been approved. These on the role of CSFs in the treatment of SCLC
developments highlight the need to reassess the current (Berghmans et al. 2002). Twelve randomized trials
evidence and to update the existing guidelines including a total number of 2,107 patients divided into
regarding the prophylactic use of G-CSF. This update three groups were considered: maintenance of dose-
was published in 2010 (Aapro et al. 2011). intensity with standard doses and time intervals of CH;
The strongest evidence supporting the use of accelerated CH with increase dose-intensity; and
G-CSF to prevent FN comes from three level I concentration of CH on an overall shorter duration time
metaanalyses (Lyman et al. 2002; Bohlius et al. 2008; with a lower number of cycles. The results of this
Kuderer et al. 2007). This last trial described the systematic review were negative for all the strategies:
information about 13 randomised trials and with 3122 in the maintenance group, CSF administration was
patients with lymphoma and solid tumor and reported associated with a detrimental effect on overall survival;
that the addition of G-CSF to standard CH resulted in in the accelerated group, no significant impact was
a significant reduction in early mortality. A small found in the response rate or survival, and concentrated
level II study has suggested a trend to improved long- CH was associated with no difference in response rate
term survival in patients with favorable-prognosis and a reduced survival.
SCLC receiving VICE CH (vincristine–ifosfamide– Only a few randomized trials have been reported
carboplatin–etoposide) plus G-CSF compared with on CH with or without granulocyte growth factors as
CH alone (Woll et al. 1995). primary prophylaxis in NSCLC. On the other hand, in
Moreover, a harmful effect has been observed with patients receiving first-line CH for advanced NSCLC,
the use of this cytokine in patients with an intratho- the occurrence of CH-induced neutropenia has been
racic stage who had been concomitantly treated with associated with a significantly longer survival
CH and radiotherapy as well as in extrathoracic stages (Di Maio et al. 2005).
treated with high-dose CH (Adams et al. 2002). In In the recent years, adjuvant CH after radical sur-
1996, the American Society of Clinical Oncology gery has become the standard for early stage NSCLC.
(ASCO) recommended that the use of CSF be avoided Cisplatin-based chemotherapy was used in all the
in patients who had received concomitant radioCH, recent trials showing a significant advantage for
and 4 years later specified that its use should be treatment compared to observation (Douillard et al.
avoided in patients with radioCH if the mediastinum 2006). Nonetheless, despite the significant hemato-
had been irradiated (Ozer et al. 2000) as in the case of logical toxicity of these regimens, the incidence of FN
LC. Nevertheless, an ongoing trial in SCL, the is reportedly much lower than 20%, therefore primary
CONVERT trial, is currently studying the possibility prophylaxis with CSFs according to guidelines is not
of CSF administration in a few cases of LC receiving advised (Winton et al. 2005).
irradiation. In relation to anemia, another known effect of bone
The use of antibiotic prophylaxis to prevent marrow toxicity, it should be remembered that its
infection and infection-related complications in can- etiology is multifactorial and includes an inappropri-
cer patients at risk of neutropenia is still contentious ate production of erythropoietin in response to the
(Cullen et al. 2005). Two metaanalysis (Gafter-Gvili alteration of the normal hemoglobin (Hb) levels. This
et al. 2005; Herbst et al. 2009) and a systematic abnormality in the production of erythropoietin is also
review (van de Wetering et al. 2005) have indicated exacerbated by CH. On the other hand, recombinant
that evidence is too limited to allow conclusions to be human erythropoietin (r-HuEPO epoietin-alfa) has
drawn regarding the relative merits of antibiotic ver- been used to improve the anemia observed in patients
sus CSF primary prophylaxis. with cancer, with an increase being observed in the
Hematological Toxicity in Lung Cancer 605

number of erythroid progenitors in both the bone In relation to thrombopenia, thrombopoietin, the
marrow and peripheral blood. synthetized factor for the stimulation of this series
Several large, community-based studies have based on preventing hemorrhagic problems after
demonstrated that epoetin-alfa effectively corrects myelosuppressive CH is still under evaluation and
anemia and improves the quality of life of anemic clinical implementation. Thrombopoietin (TPO), a
cancer patients receiving CH (Kosmidis and key physiologic regulator of platelet production, has
Krzakowski 2005). However, the contribution of been found to be the most potent thrombopoietic
erythropoietin to curative cancer treatment outcome is cytokine studied so far. Unfortunately, the clinical
controversial (Machtay et al. 2007). A safety analysis development of recombinant human thrombopoietin
in a randomized trail suggested decreased survival in has met challenges related to the biology of TPO with
patients with advanced NSCLC treated with epoeitin- a delayed peak platelet response and the findings of
alfa (Wright et al. 2007). The ASCO has made an neutralizing antibodies to the pegylated molecule
evidence level II recommendations concerning the (Vadhan-Raj 2005).
treatment of this anemia with r-Hu-EPO (Rizzo et al. In addition to the development of specific cytokines
2008). For patients with CH-associated anemia, the for the production and secretion of different hemato-
Committee continues to recommend initiating an logic cells, trials with medications such as glutation are
erythropoiesis-stimulating agent (ESA) as Hb values currently ongoing on different methods of prevention
approach or fall below 10 g/dL to increase Hb and of bone marrow toxicity. Glutation has shown to be an
decrease transfusions. effective chemoprotector against toxicity induced by
An individual patient data-analysis shows that cisplatin. Although the main experience is in ovarian
ESA increase the mortality in all patients with cancer, cancer, randomized studies in other types of tumors
and a similar increase might exist in patients on CH such as LC and the head and neck have demonstrated
(Bohlius et al. 2009). CH was given to patients in lower hematologic toxicity in patients receiving glu-
38% (72%) of 53 studies included in the metaanalysis, tation compared with the control group (Schmidinger
with radiotherapy in three (6%) and radioCH in five et al. 2000). Other drugs such as amiphostin have also
(9%). In the further five (9%) studies, no CH or shown a significant reduction in hematologic toxicity
radiotherapy was given to patients. The most frequent in randomized studies including patients with LC
tumors were breast cancer [4302 (31%) of 13933] and undergoing concurrent radioCH (Antonadou et al.
LC [3076 (22%)]. The increase in mortality seemed to 2003 and Komaki et al. 2002). Nonetheless in two
be more pronounced in patients treated with ESA more recent phase III (Movsas et al. 2005) and ran-
once a week than in those who were treated with these domized phase II studies (Han et al. 2008), both in LC,
drugs more or less often. Finally, only a few trials amiphostine was associated with higher FN.
have examined the indications for ESA that were There is a new pathway to reduce bone marrow
similar to the indications approved by the Food and toxicity secondary to radiotherapy alone or associated
Drug Administration. A study by Casas et al. (2003) with CH. Radiotherapy modulated by dose-intensity
on the approved indication also studied the impact of (IMRT) in different locations has demonstrated to be
the use of erythropoietin in the maintenance of Kar- useful in significantly reducing the doses of radio-
nofsky and Hb levels in patients with LC receiving therapy in critical tissues (Lujan et al. 2003).
con current treatment of radioCH after induction CH With IMRT planning it may be possible to reduce
(11 SCLC and 40 NSCLC). In addition to finding a both bone marrow volume at a thoracic level and
beneficial and significant impact of the administration cardiac circulation, thereby allowing blood cells to be
of erythropoietin at the level of general status and Hb irradiated with radiotherapy alone or in combination.
levels, it was also found to be a significant prognostic Prospective studies aimed at achieving a reduction in
factor of survival on multivariate analysis, together hematologic toxicity should be undertaken.
with classical factors such as weight loss and final Finally, it is currently possible to prospectively
improvement in Hb, the histology of SCLC and monitor or even predict bone marrow toxicity after
finally, Hb levels greater than 10 g/dL prior to con- radiotherapy. One article has demonstrated that the
current radioCH. variations in the cytokine called Glt-3 ligand in
606 F. Casas et al.

plasma directly reflect the damage induced by radio- Calvert AH, Ghokul S, Al Azraqui A, Wright J et al (1999)
therapy in the bone marrow during fractionated Carboplatin and paclitaxel, alone and in combination: dose
escalation, measurements of renal function, and role of the
radiotherapy, even when this damage is maintained at p53 tumor suppressor gene. Semin Oncol 26:676–684
subclinical levels (Huchet et al. 2003). This may be Cardenal F, Lopez-Cabrerizo MP, Anton A, Alberola V et al
very useful for preventive monitoring of hematologic (1999) Randomized phase III study of gemcitabine–cis-
toxicity in determined groups of patients with LC platin versus etoposide–ciisplatin in the treatment of locally
advanced or metastatic non-small-cell lung cancer. J Clin
receiving CH or radiotherapy treatment. Oncol 17:12–18
Casas F, Viñolas N, Ferrer F, Farrús B, Gimferrer JM, Agustí
C, Belda J, Luburich P (2003) Improvement in performance
References status after erythropoietin treatment in lung cancer patients
undergoing concurrent chemoradiotherapy. Int J Radiat
Oncol Biol Phys 55:116–124
Aapro MS, Bohlius J, Cameron DA et al (2011) 2010 update of Crawford J, Caserta C, Roila F (2010) Hematopoietic growth
EORTC guidelines for the use of granulocyte-colony factors ESMO clinical practice guidelines for the applica-
stimulating factor to reduce the incidence of chemotherapy- tions. Ann Oncol 21(suppl 5):v248–v251
induced febrile neutropenia in adult patients with lympho- Cullen M, Steven N, Billingham L et al (2005) Antibacterial
proliferative disorders and solid tumours. Eur J Cancer prophylaxis after chemotherapy for solid tumors and
47:8–32 lymphomas. N Eng J Med 353:988–998
Aapro MS, Cameron DA, Pettengell R et al (2006) EORTC De Souza Viana L, Serufo JC, da Costa Rocha MO et al (2008)
guidelines for the use of granulocyte-colony stimulating Performance of a modified MASCC index score for
factor to reduce the incidence of chemotherapy induced identifying low-risk febrile neutropenic cancer patients.
febrile neutropenia in adult patients with lymphomas and Support Care Cancer 16:841–846
solid tumours. Eur J Cancer 42:2433–2453 Dillman RO, Seagren SL, Propert KJ et al (1990) A randomized
Abrams RA, Lichter AS, Bromer RH, Minna JD, Cohen MH, trial of induction chemotherapy plus high-dose radiation
Deisseroth AB (1985) The hematopoietic toxicity of versus radiation alone in stage III non-small cell lung
regional radiation therapy. Correlations for combined cancer. N Engl J Med 323:940–945
modality therapy with systemic chemotherapy. Cancer Di Maio M, Gridelli C, Gallo C, Shepherd F, Piantedosi FV,
55:1429–1435 Cigolari S et al (2005) Chemotherapy-induced neutropenia
Adams JR, Lyman GH, Djubegovic B, Feinglass J et al (2002) and treatment efficacy in advanced non-small-cell lung
G-CSF as prophylaxis of febrile neutropenia in SCLC. cancer: a pooled analysis of three randomized trials. Lancet
Review of findings from 13 studies of cost-effectiveness, Oncol 6:669–677
evidence-based guidelines, patterns of care and surveys of Douillard JY, Rosell R, De Lena M et al (2006) Adjuvant
ASCO members. Expert Opin Pharmacother 3:1273–1281 vinorelbine plus cisplatin versus observation in patients with
Antonadou D, Throuvalas N, Petridis A, Bolanos N, Sagriotis completely resected stage IB–IIIA non-small-cell lung
A, Synodinou M (2003) Effect of amifostine on toxicities cancer (Adjuvant Navelbine International Trialist Associa-
with radiochemotherapy in patients with locally advanced tion [Anita]): a randomised controlled trail. Lancet Oncol
non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 7:719–727
57:402–408 Favre-Finn C, Blackhall F, Snee M, Harden S, Hulse P, Lorigan
Bennett CL, Weeks JA, Somerfield MR, Feinglass J, Smith TJ P (2010) Improving survival with thoracic radiotherapy in
(1999) Use of hematopoietic colony-stimulating factors: patients with small cell lung cancer: the CONVERT and the
comparison of the 1994 and 1997 American Society of REST trials. Clinical Oncol 22:547–549
Clinical Oncology surveys regarding ASCO clinical prac- Fukuoka M, Furuse K, Saijo N et al (1991) Randomized trial of
tice guidelines. Health Services Research Committee of the cyclophosphamide, doxorubicin, and vincristine versus
American Society of Clinical Oncology. J Clin Oncol cisplatin and etoposide versus alternation of these regimens
17:3676–3681 in small cell lung cancer. JNCI 83:885–891
Berghmans T, Paesmans M, Lafitte JJ et al (2002) Role of Furuse K, Fukuoka M, Kawahara M et al (1999) Phase III study
granulocyte and granulocyte–macrophage colony-stimulat- of concurrent versus sequential thoracic radiotherapy in
ing factors in the treatment of small-cell lung cancer: a combination with mitomycin, vindesine and cisplatin in
systematic review of the literature with methodological unresectable stage III non-small-cell lung cancer. J Clin
assessment and meta-analysis. Lung Cancer 37: Oncol 17:2692–2699
115–123 Gafter-Gvili A, Fraser A, Paul M, Leibovici L (2005) Meta-
Bohlius J, Herbst C, Reiser M, Schwarzer G, Engert A. (2008) analysis:antibiotic prophylaxis reduces mortality in neutro-
Granulopoiesis –stimulating factors to prevent adverse penic patients. Ann Intern Med 142.977-95
effects in the treatment of malignant lymphoma (Review). Gregor A, Drings P, Burghouts J, Postmus PE, Morgan D,
Cochrane Database Syst Rev (4):CD003189, Oct 8. Sahmoud T (1997) Randomized trial of alternating versus
Bohlius J, Scmindlin K, Brillant C, Schwarzer G, Trelle S et al sequential radiotherapy/chemotherapy in limited-disease
(2009) Recombinant human erythropoiesis-stimulating patients with small-cell lung cancer: a European Organiza-
agents and mortality in patients with cancer: a meta- tion for Research and Treatment of Cancer Lung Cancer
analysis of randomized trials. Lancet 373:1532–1542 Cooperative Group Study. J Clin Oncol 15:2840–2849
Hematological Toxicity in Lung Cancer 607

Han HY, Han J-Y, Yu SY, Pyo HR, Kim HY, Cho KH, Lee DH Le Chevalier T, Arraigada R, Quoix E et al (1991) Radiother-
et al (2008) Randomized phase 2 study of subcutaneous apy alone versus combined chemotherapy and radiotherapy
amifostine versus epoetin-alfa given 3 times weekly during in nonresectable non-small-cell lung cancer. First analysis
concurrent chemotherapy and hyperfractionated radiother- of a randomized trial in 353 patients. J Natl Cancer Inst
apy for limited-disease small cell lung cancer. Cancer 83:417–423
113:1623–1631 Lujan AE, Mundt AJ, Yamada SD, Rotmensch J, Roeske JC
Hanna N, Neubauer M, Yiannoutos C, McGarry R, Arsenau J, (2003) Intensity-modulated radiotherapy as a means of
Ansari R et al (2008) Phase III study of cisplatin, etoposide, reducing dose to bone marrow in gynecologic patients
and concurrent chest radiation with or without consolidation recieving whole pelvic radiotherapy. Int J Radiar Oncol Biol
docetaxel in patients with inoperable stage III non-small- Phys 57:515–521
cell lung cancer: the Hoosier Oncology Group and US Luo J, Lear SJ, Xu Y, Zheng D (2010) Comparison of cisplatin
oncology. J Clin Oncol 35:5755–5760 and carboplatin based third-generation chemotherapy in
Herbst C, Naumann F, Kruse EB et al. (2009). Prophylactic 1014 Chinese patients with advanced non-small-cell lung
antibiotics or G-CSF for the prevention of infections and cancer. Med Oncol (in press)
improvement of survival in cancer patients undergoing Lyman GH, Kuderer NM, Djulbegovic B (2002) Prophylactic
chemotherapy. Cochrane Database Syst Rev (1):CD007107, granulocyte colony-stimulating factor in patients receiving
Jan 21 dose-intensive cancer chemotherapy: a meta-analysis. Am J
Huber RM, Flentje M, Scmindt M, Pöllinger B, Gosse H, Med 112:406–411
Wilner J (2006) Simultaneous chemoradiotherapy compared Machtay M, Pajak TF, Suntharalingam M et al (2007)
with radiotherapy alone after induction chemotherapy in Radiotherapy with or without erythropoietin for anemic
inoperable stage IIIA or IIIB non-small-cell lung cancer patients with head and neck cancer: a randomized trial of
study CTR 799/97 by the Bronchial Carcinoma Therapy the Radiation Therapy Oncology Group (RTOG 99–03). Int
Group. J Clin Oncol 27:4397–4404 J Radiat Oncol Biol Phys 69:1008–1017
Huchet A, Belkacemi Y, Frick J, Prat M et al (2003) Plasma Milward MJ, Boyer MJ, Lehnert M et al (2003) Docetaxel and
Flt-3 ligand concentration correlated with radiation-induced carboplatin is an active regimen in advanced non-small-cell
bone marrow damage during local fractionated radiother- lung cancer: a phase II study in Caucasian and Asian
apy. Int J Radiat Oncol Biol Phys 57:508–515 patients. Ann Oncol 14:449–454
Jeremic B, Acimovic L, Djuric L (1995) Randomized trial of Movsas B, Scott C, Langer C et al (2005) Randomized trial of
hyperfractionated radiation therapy with or without con- amifostine in locally advanced non-small-cell lung cancer
current chemotherapy for stage III non-small-cell lung patients receiving chemotherapy and hyperfractionated
cancer. J Clin Oncol 13:452–458 radiation: Radiation Therapy Oncology Group trial 98-01.
Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1997) J Clin Oncol 23(10):2145-2154
Initial versus delayed accelerated hyperfractionated radiation Murray N, Coy P, Pater JL, Hodson I et al (1993) Importance of
therapy and concurrent chemotherapy in limited small-cell timing for thoracic irradiation in the combined modality
lung cancer: a randomized study. J Clin Oncol 15:893–900 treatment of limited-stage SCLC. J Clin Oncol 11:336–344
Kelly K, Crowley J, Bunn PA, Presant CA et al (2001) Ozer H, Armitage JO, Bennet Ch L et al (2000) 2000 update of
Randomized phase III trial of paclitaxel plus carboplatin recommendations for the use of hematopoietic colony-
versus vinorelbine plus cisplatin in the treatment of patients stimulating factors: evidence-based, clinical practice guide-
with advanced non-small-cell lung cancer: a Southwest lines. J Clin Oncol 18:3558–3585
Oncology Group trial. J Clin Oncol 19:3210–3218 Pirker R, Pereira JR, Szczesna A et al (2009) Cetuximab plus
Khan S, Dhadda A, Fyfe D, Sundar S (2008) Impact of chemotherapy in patients with advanced non-small-cell lung
neutropenia on delivering planned chemotherapy for solid cancer (FLEX): an open-label randomized phase III trial.
tumours. Eur J Cancer Care 17:19–25 Lancet 373:1525–1531
Klastersky J, Awada A, Paesmans M, Aoun M (2011) Febrile Pivot X, Guardiola E, Etienne M et al (2000) An analysis of
neutropenia: a critical review of the initial management. potential factors allowing an individual prediction of
Crit Rev Oncol Hematol 78(3):185-194 cisplatin-induced anemia. Eur J Cancer 36:852–857
Komaki R, Lee JS, Kaplan B et al (2002) Randomized phase III Radosavljevic D, Golubcic I, Gavrilovic D, Kezic I, Jelic S
study of chemoradiation with or without amifostine for (2009) Do the time to chemotherapy response and the dose
patients with favorable performance status inoperable stage intensity have an impact on patient outcome in advanced
I–III non-small cell lung cancer: preliminary results. Semin non-small cell lung cancer? J BUON 14:203–209
Radiat Oncol 12:46–49 Ratain MJ, Schilsky RL, Conley BA, Egorin MJ (1990)
Kosmidis P, Krzakowski M (2005) ECAS Investigators. Pharmacodynamics in cancer therapy. J Clin Oncol
Anemia profiles in patients with lung cancer: what have 8:1739–1753
learned from the European Cancer Anaemia Survey Reck M, von Pawel J, Zatlooukal P et al (2009) Phase III trial
(ECAS)? Lung Cancer 50:401–412 of clisplatin plus gemcitabine with either placebo or
Kuderer NM, Dale DC, Crawford J, Lyman GH (2007) Impact bevacizumab as first-line therapy fro nonsquamous non-
of primary prophylaxis with granulocyte colony-stimulating small-cell lung cancer: AVAIl. J Clin Oncol 27:1227–1234
factor on febrile neutropenia and mortality in adult cancer Rizzo JD, Somerfiweld MR, Hagerty KL, Seindenfeld J,
patients receiving chemotherapy: a systematic review. Bohlius J et al (2008) Use of epoetin and darbopetin in
J Clin Oncol 25:3158–3167 patients with cancer: 2007 American Society of Clinical
608 F. Casas et al.

Oncology/American Society of Hematology Clinical Prac- Tubiana M, Frindel E, Croizat H (1979) Effects of radiation on
tice Guideline Update. J Clin Oncol 26:132–149 bone marrow. Pathol Biol 27:326–334
Sandler A, Gray R, Perry MC et al (2006) Placlitaxel– Turrisi AT, Kim K, Blum R et al (1999) Twice-daily compared
carboplatin alone or with bevacizumab for non-small-cell with once-daily thoracic radiotherapy in limited small-cell
lung cancer. N Engl J Med 355:2542–2550 lung cancer treated concurrently with cisplatin and etopo-
Saunders MI, Mauguen A, Le Pechoux C, Schild S, Turrisi et al side. N Engl J Med 340:265–271
(2010) Meta-analysis of modified fractionation radiotherapy Vadhan-Raj S (2005) Thrombopoietic growth factors and
in lung cancer on 9 randomized trials. In: Proceedings cytokines. Curr Hematol Rep 4:137–144
ESTRO 29 Barcelona, p 29 Van de Wetering MD, de Witte MA, Kremer LC et al (2005)
Sause WT, Scott C, Taylor S et al (1995) Radiation Therapy Efficacy of oral prophylactic antibiotics in neutropenic
Oncology Group (RTOG) 88-08 and Eastern Cooperative afebrile oncology patients: a systematic review of random-
Oncology Group (ECOG) 4588: preliminary results of a ised controlled trials. Eur J Cancer 41:1372–1382
phase III trial in regionally advanced, unresected non-small Vokes EE, Herdon JE, Kelley MJ, Cichetti MG et al (2007)
cell lung cancer. J Natl Cancer Inst 87:198–205 Induction chemotherapy followed by chemoradiotherapy
Schaake-Koning C, van der Bogaert W, Dalesio O et al (1992) compared with chemoradiotherapy alone for regionally
Effects of concomitant cisplatin and radiotherapy on advanced unresectable stage III NSCLC: Cancer and
inoperable non-small-cell lung cancer. N Engl J Med leucemia Group B. J Clin Oncol 13:1698–1704
326:524–530 Vokes EE, Rendón JE, Crawford J, Leopold KA, Perry MC
Schiller JH, Harrington D, Belani CP et al (2002) Comparison et al (2002) Randomized phase II study of cisplatin with
of four chemotherapy regimens for advanced non-small-cell gemcitabine or placlitaxel or vinorelbine as induction
lung cancer. N Eng J Med 346:92–98 chemotherapy followed by concomitant chemoradiother-
Schmidinger M, Budinsky AC, Wenzel C, Piribauer M et al apy for stage IIIB non-small-cell lung cancer: cancer
(2000) Glutathione in the prevention of cisplatin induced and leukemia group B study 9431. J Clin Oncol 20:
toxicities. A prospectively randomized pilot trial in patients 4191–4198
with head and neck cancers and non small cell lung cancer. Winton T, Livingston R, Johnson D, Rigas J et al (2005)
Wien Klin Wochenschr 28:617–623 Vinorelbine plus cisplatin vs. observation in resected non-
Takada M, Fukuoka M, Kawahara M et al (2002) S. Phase III small-cell lung cancer. N Eng J Med 352:2589–2597
Study of concurrent versus sequential thoracic radiotherapy Woll PJ, Hodgetts J, Lomax L et al (1995) Can cytotoxic dose-
in combination with cisplatin and etoposide for limited- intensity be increased by using granulocyte colony-stimu-
stage small-cell lung cancer: results of the Japan Clinical lating factor? A randomized controlled trial of lenograstrim
Oncology Group study 9104. J Clin Oncol 20:3054–3060 in small-cell lung cancer. J Clin Oncol 13:652–659
Timmer-Bonte JN, de Boo TM, Smith HJ et al (2005) Wright JR, Ung YC, Julian JA et al (2007) Randomized,
Prevention of chemotherapy-induced febrile neutropenia double-blind, placebo-controlled trial of erythropoietin in
by prophylactic antibiotics plus or minus granulocyte non-small-cell lung cancer with disease-related anemia.
colony-stimulating factor in small-cell lung cancer: a Dutch J Clin Oncol 25:1027–1032
randomized phase III study. J Clin Oncol 23:7994–84 Zatloukal P, Petruzelka L, Zemanova M, Havel L, Janku F,
Trovo MG, Minatel E, Franchin G, Boccieri MG et al (1992) Judas L et al (2004) Concurrent versus sequential chemo-
Radiotherapy versus radiotherapy enhanced by cisplatin in radiotherapy with cisplatin and vinorelbine in locally
stage III non-small cell lung cancer. Int J Radiat Oncol Biol advanced non-small cell lung cancer: a randomized study.
Phys 254:11–15 Lung Cancer 46:87–98
Radiation-Induced Lung and Heart Toxicity
Liyi Xie, Xiaoli Yu, Zeljko Vujaskovic, Mitchell S. Anscher,
Timothy D. Shafman, Keith Miller, Robert Prosnitz,
and Lawrence Marks

Contents 4 Modifiers of RT-Induced Lung Injury ................. 616


5 Cardiotoxic Effects of Thoracic Radiation
1 Pulmonary Effects of Thoracic Radiation Therapy..................................................................... 617
Therapy..................................................................... 609 References.......................................................................... 620
1.1 Clinical RT-Induced Lung Toxicity ......................... 609
2 Biology of Radiation-Induced Lung Injury.......... 612
Abstract
3 Predictors of RT-Induced Lung Injury ................ 614
For many patients with lung cancer, thoracic
radiation therapy (TRT) is an integral part of their
treatment. The effect of TRT on normal structures
L. Xie  L. Marks (&)
Department of Radiation Oncology, is an important consideration when optimizing
University of North Carolina, Campus Box 7512, treatment plans for patients. This chapter will
101 Manning Drive, Chapel Hill, NC 27514, USA review radiation therapy (RT)-induced lung and
e-mail: marks@med.unc.edu
heart injury including both the clinical and
L. Xie  X. Yu biological mechanisms for these toxicities. It
Department of Radiation Oncology,
will also analyze the variety of predictors of
Fudan University Shanghai Cancer Center,
200032 Shanghai, China RT-induced lung and heart damage as well as
methods to prevent and treat these toxicities.
Z. Vujaskovic
Radiation Oncology and Pathology,
Department of Radiation Oncology,
Duke University Medical Center, Box 3455,
Durham, NC 27710, USA
M. S. Anscher
1 Pulmonary Effects of Thoracic
Department of Radiation Oncology, Radiation Therapy
Virginia Commonwealth University School of Medicine,
401 College Street, PO Box 980058, 1.1 Clinical RT-Induced Lung Toxicity
Richmond, VA 23298-0058, USA
T. D. Shafman 1.1.1 Introduction
Landmark Medical Centre, 115 Cass Ave,
Radiation-induced lung toxicity is a common occur-
Woonsocket, RI 02895, USA
rence in patients treated with curative intent for
K. Miller
lung cancer. Approximately 5–20% of patients
21st Century Oncology, Fort Myers, FL, USA
treated with RT for lung cancer have been reported to
R. Prosnitz
develop RT-induced lung injury (Favaretto et al.
Department of Radiation Oncology,
University of Pennsylvania School of Medicine, 1996; Segawa et al. 1997; Fu et al. 1997; Monson et al.
2 Donner Building, 3400 Spruce Street, 1998; Yamada et al. 1998; Nyman et al. 1998;
Philadelphia, PA 19104, USA

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 609
DOI: 10.1007/174_2011_273, Ó Springer-Verlag Berlin Heidelberg 2011
610 L. Xie et al.

Table 1 Incidence of RT-induced clinical and radiologic lung injury


Reference (No) Number of Symptom rate (%) Incidence of radiologic Follow-up duration
patients Grade Grade Grade Grade changes (%) (months)
1 2 3 4
Favaretto et al. 39 13 64–90
(1996)
Segawa et al. 89 38a 5.6 8.9 5.6 58 –
(1997)
Fu et al. (1997) 60 – 17a 8 – 23
Monson et al. 83 8 2 7 34 C4
(1998)
Yamada et al. 60 – 13.3a 11.6 3.4 85 C6
(1998)
Nyman et al. 90 – – 8a – 22 (minimum)
(1998)
Van den Brande 23 – – 11a – [24
et al. (1998)
Makimoto et al. 111 15 \6
(1999)
Graham et al. 99 – 14–20a 24 (median)
(1999)
Robnett et al. 144 – – 8.3a 11.5 (median)
(2000)
Sunyach et al. 54 – 29 (Lent-Soma scale) 37 C6
(2000)
Inoue et al. (2001) 191 36a 13 C12
Hernando et al. 201 – 13 4 – C6
(2001)
Oetzel et al. (1995) 46 20 [3
Perry et al. (1987) 391 7 17.3 (median)
Gross (1977) _ 5–15 65 _
Simpson et al. 316 3 36
(1985)
Kwon et al. (2000) 47 – 8.5a – – 2–33
Perez et al. (1980) 365 4 60
Martel et al. (1994) 42 21.4 [7
Tucker et al. (2010) 442 14.9a 4.5 22.2 2.5 C6
– 22.6b 21 0.5
a
RTOG criteria
b
CTCAE 3.0

Van den Brande et al. 1998; Makimoto et al. 1999; chronic pulmonary dysfunction requiring oxygen
Graham et al. 1999; Robnett et al. 2000; Sunyach et al. therapy and potentially leading to death. The wide
2000; Inoue et al. 2001; Hernando et al. 2001; Oetzel range in incidence of reported toxicity is secondary to
et al. 1995; Perry et al. 1987; Gross 1977; Simpson the different methods used to measure pulmonary
et al. 1985; Kwon et al. 2000; Perez et al. 1980; Martel dysfunction. While most patients have radiographic
et al. 1994; Tucker et al. 2010) (Table 1). Clinical changes, few have changes in functional endpoints and
symptoms range from mild shortness of breath to even less have severe clinical symptoms (Table 1).
Radiation-Induced Lung and Heart Toxicity 611

Clinical endpoints for RT-induced lung injury have corticosteroids, and in some cases, supplemental
traditionally been divided into acute (early) and oxygen is needed. Similar to acute pneummonitis,
chronic (late) toxicity. Acute pneumonitis typically tumor progression, infection and COPD flare must be
occurs 1–6 months after TRT, with a peak incidence ruled out as exacerbating factors for symptomatic
of around three months (Van Dyk et al. 1981). fibrosis and treated appropriately. As noted earlier,
Chronic lung fibrosis usually evolves six months to radiographic evidence of regional lung scarring is seen
several years after treatment. in almost all patients, including those without clinical
symptoms. There does not appear to be an association
1.1.2 Early Toxicity between the presence of an abnormality on CT scan
Patients with RT-induced pneumonitis often present and the development of symptoms, though this has not
shortness of breath, cough, congestion, and some may been extensively studied. (Garipagaoglu et al. 1999).
have a low-grade fever. For patients with chronic After high doses of TRT there have been rare reports
obstructive pulmonary disease (COPD), it may be of pulmonary complications, such as bronchial
difficult to distinguish a flare COPD from acute stenosis, bronchomalacia, and mediastinal fibrosis
pneumonitis. In general, the severity of the symptoms with secondary recurrent laryngeal nerve injury
is related to the amount of normal lung irradiated. (Maguire et al. 2001; Dechambre et al. 1998).
Pneumonitis usually responds well to steroids and
40–60 mg of prednisone each day for several weeks, 1.1.4 Radiographic Changes
followed by a slow taper. This provides relief for most Radiographic findings are common in patients fol-
patients. It is important to consider the possibility of lowing TRT, and also among patients who do not
infection since this might become worse due to the have symptoms of RT-induced lung injury. The fre-
steroids. In situations where either infection or quency of finding these changes depends on the type
pneumonitis appear likely, an initial trial of empiric of radiographic assessment performed. A chest x-ray
antibiotics, followed by steroids if there is no (CXR) performed after TRT can reveal a diffuse
response to antibiotics, may be indicated. In patients infiltrate corresponding to the radiation field. There
with an unsatisfactory response to either treatment, can also be an associated volume loss of the affected
tumor progression and/or lymphangitic tumor should portion of the lung and in late toxicity, there can be
be considered. Severe RT-induced pneumonitis can extension of the findings outside the treated area and
result in severe respiratory distress requiring hospi- deviation of the trachea towards the irradiated area.
talization, intubation and possibly death. Computed tomography (CT) scans are more sen-
Thoracic radiation therapy can also cause acute sitive than CXR and can detect abnormalities in
irritation of the pleura, with secondary pleuritic pain more than 50% of patients (Mah et al. 1987). CT
and this can be treated with anti-inflammatory and/or scans are very sensitive to slight changes in lung
narcotic pain relief medicines. Irritation of the trachea densities and therefore are the favored diagnostic
and bronchial airways, which can lead to a cough, procedure for the detection of RT-induced lung
may also occur and is treated with cough suppression injury (Mah et al. 1987; Libshitz and Shuman 1984).
medicines. There is a well-defined dose–response relationship
for the patterns seen on CT scans after TRT and
1.1.3 Late Toxicity these include: homogeneous, slight increase in lung
The most prominent late consequence of TRT is pul- density; patchy consolidation; discrete consolidation;
monary fibrosis. Radiological changes consistent with solid consolidation (Libshitz and Shuman 1984;
fibrosis are seen in most patients. Symptomatic Mah et al. 1986). Chronic changes in the thorax that
patients show progressive chronic dyspnea and this can be seen on the CT scan following TRT include
can occur months to years after TRT (Gross 1977; lung contraction, pleural thickening, tenting of the
Perez et al. 1980; Martel et al. 1994; McDonald et al. diaphragm, and deviation of the mediastinal struc-
1995; Morgan and Breit 1995; Abid et al. 2001). tures toward the treated area. These can appear
Relief from symptoms is the goal of treatment as months to years after radiotherapy.
the reversal of the fibrosis is highly unlikely. Treat- Lung perfusion and ventilation can be abnormal
ment includes anti-inflammatory agents such as following TRT (Gross 1977; Prato et al. 1977).
612 L. Xie et al.

Single-photon-emission computed tomography (RV). The volume of air in the lung can increase
(SPECT) perfusion and ventilation scans are more following TRT if there is an expansion of lung
frequently abnormal than planar images, similar to volume or could decrease secondary to fibrosis.
CT’s advantage over chest X-rays (CXR) (Gross Reports in the literature describe both and the range
1977; Prato et al. 1977). Perfusion appears to be more varies from -20% to +9.5% (Van den Brande et al.
sensitive than ventilation in the evaluation of RT- 1998; Sunyach et al. 2000; Mattson et al. 1987;
induced lung injury, and both are more sensitive than Choi and Kanarek 1994; Rubenstein et al. 1988;
CXR or CT (Bell et al. 1988; Shapiro et al. 1990). Brady et al. 1965; Bonnet et al. 2001) (Table 2).
This is most apparent at modest doses, 15–40 Gy, Gas exchange in the lungs is measured via the
where often there is no change seen in tissue density, carbon monoxide (CO) diffusion capacity (DLCO)
yet clear reductions in both ventilation and perfusion. quantifying the transfer of CO from inspired gas into
Perfusion and ventilation abnormalities have been pulmonary capillary blood. Many complex factors,
seen in 53–95 and 35–45% of irradiated patients, besides gas diffusion, contribute to DLCO, including
respectively. The inconsistencies between changes in ventilation/perfusion characteristics of alveolar units,
ventilation and perfusion support the idea that capillary blood volume, hemoglobin concentration,
following TRT, some areas remain ventilated, but are and the reaction rates between CO and hemoglobin.
not adequately perfused (Bell et al. 1988; Marks et al. In addition, other clinical factors such as diurnal
1993). variation, menstrual cycle, ethanol ingestion, and
Several studies that have reported rates of both cigarette smoking can effect DLCO (Garipagaoglu
clinical pneumonitis and radiographic abnormalities et al. 1999). The DLCO is frequently corrected for
following TRT are summarized in Table 1. As shown, anemia but the other factors known to affect the
radiographic changes occur far more frequently than DLCO are difficult to control during its calculation.
clinical symptoms. (Garipagaoglu et al. 1999). The DLCO tends to be
affected by TRT to a greater degree than the other
1.1.5 Functional Endpoints parameters and has been shown to be reduced from 5
In general, abnormalities in pulmonary function tests to 35% following TRT (Table 2). It is difficult to
(PFTs) do not occur during the first weeks following generalize changes in PFTs, given the wide variety of
TRT. Following this, changes in PFTs can occur pre-treatment values and the diverse amounts of lung
along with the signs and symptoms of pneummonitis irradiated in each patient. Other standard measure-
and/or fibrosis. Pulmonary function tests measure the ments of pulmonary function, such as the 6 min walk
transfer of large volumes of air through the conduct- test or exercise stress tests, have not been routinely
ing airways, and transfer of gases through the alveolar used to measure RT-induced lung injury.
surfaces. Spirometry assesses the rate of gas move-
ment and the most commonly measured parameter is
the forced expiratory volume in one second (FEV1). 2 Biology of Radiation-Induced Lung
FEV1 is a measurement of air movement and can be Injury
normalized to the forced vital capacity (FVC), a
measurement of ‘‘useful’’ lung volume, which is the Radiation-induced lung injury is characterized by
FEV1% (FEV1/FVC). While the response of the progressive histological changes which are linked
tumor to TRT may lead to an increase in FEV1, FVC with the clinical symptoms and radiological findings
may decrease secondary to restrictive disease (fibro- of pulmonary dysfunction. Current thinking suggests
sis) and thus FEV1% (FEV1/FVC) may increase. that the underlying molecular processes of RT-
Reductions in FEV1 following TRT range from 0 to induced lung injury is a dynamic interplay of several
30%, but there are a wide variety of confounding molecular factors. These processes can be loosely
variables that limit the meaningful interpretation of separated into three major phases.
this data. The early responses to pulmonary radiation include
A variety of PFTs measure lung volume including an increased production of reactive oxygen/nitrogen
the total lung capacity (TLC), vital capacity (VC), (ROS/RNS) species and proinflammatory/profibrotic
forced vital capacity (FVC), and residual volume cytokine expression resulting in tissue inflammation.
Radiation-Induced Lung and Heart Toxicity 613

Table 2 Percent reduction in pulmonary function parameters after thoracic radiation therapy
Reference (No) FEV1 DLCO FVC TLC VC
Sunyach et al. (2000) +0.1 4.3 6.5
Marks et al. (1993) 27 16
Van den Brande et al. (1998) 10 25 15
Mattson et al. (1987) 18 28 22 10 20
Choi and Kanarek (1994) 11
Rubenstein et al. (1988) 14
Brady et al. (1965) 8 11.5 5.5 +6

Simultaneously, DNA damage in various cell types, provoke the cell loss and aggravate hypoxia
including type I and II pneumocytes and endothelial throughout the transition to late toxicities. There is
cells, results in apoptosis, which further triggers the progressive fibrosis of alveolar septae, which become
secretion of growth factors and proteases, and deg- thickened with bundles of elastic fibers while small
radation of the extracellular matrix. This process also vessel walls become obliterated by collagen deposits
leads to increased permeability and loss of integrity of (Gross 1977; Katzenstein and Askin 2006; Roswit and
vessels, leading to edema, impaired tissue perfusion, White 1977). The alveoli eventually collapse and
and hypoxia. The initial latent period following TRT become obliterated by connective tissue. This usually
until the manifestation of detectable injury may reflect occurs beyond 4–6 months following TRT. This is an
the inherent turnover time of these cells (Gross 1977; active process and there is evidence that it may be
Travis et al. 1977; Travis 1980). genetically determined (Haston et al. 2002).
Histologically, RT-induced pneumonitis is typified Many of the molecular events described above
by an exudate of proteinaceous material into the occur as a result of increased expression and activa-
alveoli, desquamation of epithelial cells from the tion of transcription factors that regulate oxidative
alveolar lining, alveolar edema, and an infiltration of stress response and cytokine function (Brach et al.
inflammatory cells. This leads to thickening of the 1991; Hallahan et al. 1991, 1994). After exposure to
alveolar septa, reduced lung compliance, and even- ionizing radiation these events proceed rapidly and
tually impairment of gas exchange. Radiation causes involve multiple cells within the lungs (Hong et al.
the early release of surfactant by type II pneumocytes 1995, 1997; Rubin et al. 1995). These events occur
and this results in alterations in alveolar surface ten- during a period of clinically normal lung function,
sion and low lung compliance (Rubin et al. 1980, and result in elevated tissue levels of ROS/RNS,
1983). proinflammatory/profibrotic cytokines, such as IL-1,
As noted above, damage to vascular endothelial TNF-a PDGF, and TGF-b (Rubin et al. 1995;
cells results in increased permeability of capillaries Finkelstein et al. 1994; Franko et al. 1997; Epperly
(Gross 1977). The endothelial cells become pleo- et al. 1999; Hallahan et al. 1989). If sustained, this
morphic, vacuolated, producing areas of denuded chronic inflammation and overexpression of cytokines
basement membrane and occlusion of the capillary lead to overt lung injury.
lumen by debris and thrombi (Gross 1980). Many of Late injury is often observed in the form of fibro-
these findings are apparent well before RT-induced sis. Histologically, lung fibrosis and pulmonary
pneumonitis develops and they persist throughout the hypoxia can be seen by about six months post-RT,
course of the illness and beyond. and are characterized by increased collagen and
A delayed response, consisting of a second wave expression of the hypoxia marker carbonic anhydrase-
of cytokine induction and hypoxia along with mac- 9. The chronic overexpression of ROS/RNS and
rophage infiltration in the lung, occurs at 6–8 weeks. cytokines maintains the chronic inflammatory state
Chronic inflammation, as a result of the recruitment and contributes to the progression of RT-induced lung
of leukocytes and cascade of chemokines, cytokines, fibrosis (Johnston et al. 2002) [for details see the
and growth factors at the injury site, can further recent review by Graves et al. (2010)].
614 L. Xie et al.

studies demonstrate that these ‘‘threshold’’ dosimetric


3 Predictors of RT-Induced Lung parameters are useful in predicting the likelihood of
Injury RT-induced lung injury. Similarly, the mean lung dose
has also been associated with RT-induced lung injury.
Given the pitfalls of diagnosing and describing the The recent QUANTEC review provides a good
continuum of clinical and radiological RT-induced summary of the available data relating the risk of
lung damage, predicting its occurrence is complicated symptomatic pneumonitis to either threshold doses, or
and fraught with deficiencies. The quality of the mean lung dose in Figure 2 of that paper. Note the
predictions is related to the endpoint chosen and the gradual increase in pneumonitis risk with mean dose
method used to calculate the risk. (Marks et al. 2010). While these parameters have been
Several studies tried to relate changes in PFTs individually correlated with clinically significant lung
based on the percent of functional lung irradiated injury, they are highly related to each other and none
(Choi and Kanarek 1994; Rubenstein et al. 1988; has been shown to be superior. The greatest benefit of
Abratt et al. 1990; Curran et al. 1992). In these these data may not be for absolute risk assessment, but
investigations, the percent of lung at risk was to provide a means to compare treatment plans for their
approximated from planar ventilation and perfusion relative risks.
scans. The observed decline in PFTs was typically It is clear from the wide variety of results that the
less than the models predicted (Choi and Kanarek volume of irradiated lung may not be sufficient to
1994; Rubenstein et al. 1988; Abratt et al. 1990; accurately predict RT-induced lung toxicity. The data
Curran et al. 1992). Consequently, investigators have derived from DVHs disregard all spatial information
used newer three-dimensional (3D) planning software and it is known that some regions of the lung have
and related local RT doses to lung SPECT perfusion/ greater functional importance. In patients with heal-
ventilation-defined regional lung injury (Woel et al. thy lungs, the ventilation perfusion ratio reveals that
2002; Seppenwoolde et al. 2000; Mah et al. 1994). gas exchange is better at the lung bases than at the
Clear dose–response relationships for radiographic apices. For lung cancer patients with COPD,
lung injury have been found. However, predictions of emphysema preferentially affects the apical lung and
PFT changes and clinical symptoms based on regional therefore the lung bases may be more important for
dose–response data have been inconsistent (Fan et al. respiration in these patients. Finally, tumor-related
2001; Theuws et al. 1998). lung dysfunction is also related to lung anatomy and
Patients with lung cancer are typically treated with is not accounted for in DVHs. Taken together, these
multiple beams that enter the lungs from different data suggest that the utility of traditional DVHs to
directions resulting in a complicated 3D dose distri- predict RT-induced lung injury may be suboptimal.
bution. Attempts to predict RT-induced lung injury Some recent studies of RT-induced lung injury uti-
from field size and dose are made difficult by an lized anatomic information and report that treatment
incomplete understanding of complex dose and volume to the lower portion of the lung may be more toxic
parameters. While both higher dose per fraction and than treatment of the upper lung, however, this has
total dose were found to be correlated with symptom- not been confirmed (Graham et al. 1999; Yorke et al.
atic lung injury, less consistent results have been found 2002a; Tsujino et al. 2003). A SPECT perfusion scan
with two-dimensional (2D) field size (Robnett et al. is able to define functional areas of the lung and
2000; Byhardt et al. 1993; Roach et al. 1995). The use therefore dose-function (i.e. perfusion) data extracted
of 3D treatment planning has provided investigators from this test may be more predictive for RT-induced
with the tools to better evaluate the risk of RT-induced lung injury than the traditional DVH (Woel et al.
lung injury. Traditionally, the 3D dose distributions are 2002; Seppenwoolde et al. 2000; Lind et al. 2002).
recalculated into a 2D dose-volume histogram (DVH) Many studies have addressed the role of potential
which is easier to interpret. The percent of lung volume biologic predictors of RT-induced lung injury. These
receiving equal to or greater than a specific dose can be are markers found in the blood prior to or during TRT
found from a DVH and typically a ‘‘single value of that reflect a predisposition for RT-induced lung
merit’’ is derived from the DVH, such as the percent of injury. TGF-b is a multifunctional regulator of cell
lung receiving at least 20 (V20) or 30 (V20) Gy. Many growth and differentiation that stimulates connective
Radiation-Induced Lung and Heart Toxicity 615

tissue formation and decreases collagen degradation with more modern agents (e.g. docetaxel and gemcit-
which can result in fibrosis. In a series of patients abine) used in concurrent/consolidate chemoradiation
receiving TRT it was found that elevated TGF-b modalities, higher rates of pulmonary toxicity may be
levels at the completion of TRT was associated with a observed (Marks et al. 2010; Barriger et al. 2010).
significantly higher incidence of clinical pneumonitis Tumor location may be a valuable component for
(Anscher et al. 1994). The dosimetric predictor V30 assessing the probability of RT-induced lung injury.
combined with the TGF-b plasma concentration has Reduction in the size of an obstructing tumor may
been shown to improve the accuracy of predicting improve respiratory status, even if some lung is
pneumonitis (Fu et al. 2001). In patients with a injured, so the prediction of post-RT lung function
V30 \ 30% and stable TGF-b during RT, the inci- can be complicated by anatomy. It has been shown
dence of symptomatic RT-induced lung injury was that patients with central obstructing tumors which
6.9%. Patients with a V30 C 30% or a TGF-b result in a shift of ventilation or perfusion away from
increasing during RT (but not both), had an incidence the area to be treated are more likely to have an
of RT-induced lung injury that was 22.8%. With a improvement of lung function following TRT (Choi
rising TGF-b and V30 C 30%, the incidence was and Kanarek 1994; Marks et al. 2000). Among
42.9% (P = 0.02). A more recent study from the patients with a V/Q shift of [10% to the uninvolved
same group (Evans et al. 2006) still showed some side of the lung by a central cancer, pulmonary
predictive ability with TGF-b in patients with high function improved in 60% of patients after RT, 20%
V30. Other cytokines have also been implicated in remained essentially stable, and only 20% had the
RT-induced lung injury. Elevated plasma levels of the reduction in PFTs that was predicted by the volume of
pro-inflammatory cytokines IL-1a and IL-6 are asso- lung irradiated (Choi and Kanarek 1994). A separate
ciated with the development of pneumonitis (Chen study demonstrated that, in patients with central lung
et al. 2002). These studies suggest that biologic tumors, 8/20 (40%) with adjacent SPECT hypoper-
markers may be useful in identifying patients at risk fusion had improvements in DLCO following radia-
for RT-induced lung injury (Zhao et al. 2009). tion, while only 3/17 (18%) of patients without
Many commonly used chemotherapeutic drugs hypoperfusion had improvement (P = 0.10) (Marks
have lung toxicity when used alone (Abid et al. 2001). et al. 2000). Patients with central tumors are likely at
The use of combinations of chemotherapy with RT, greater risk of bronchial injury following high-dose
either concurrently or sequentially, raises the concern RT (e.g. [73 Gy) than are patients with more
for added toxicity. While there is evidence for an peripherally placed lesions (Miller et al. 2005).
increased risk of pulmonary toxicity with concurrent While it is reasonable to associate cigarette smok-
RT and doxorubicin, mitomycin-C, cyclophospha- ing with an increased risk of RT-induced lung toxicity,
mide, and bleomycin, these drugs are not commonly the data are somewhat confounding. Chronic lung
used in lung cancer patients (McDonald et al. 1995). disease caused by a long history of smoking makes
Recent trials using platinum-based regimens have not patients more susceptible to lung injury, however,
shown increased RT-induced lung toxicity. In a study some data suggest that active smoking may have a
comparing induction chemotherapy with cisplatin, protective effect (Hernando et al. 2001; Garipagaoglu
vinblastine, and followed by thoracic RT to the same et al. 1999; Johansson et al. 1998). A retrospective
dose of RT alone, the frequency of severe lung toxicity review of patients with RT-induced symptomatic
was reported to be only one percent in each treatment pneumonitis following treatment of esophageal and
group (Dillman et al. 1990). In comparison of breast cancer found a lower incidence of lung injury in
sequential cisplatin, vindesine, and mitomycin with RT smokers (Johansson et al. 1998). A large study of
versus concurrent treatment with the same agents, the patients irradiated for lung cancer noted a similar
rate of grade two or higher pulmonary toxicity was finding (Jin et al. 2009). A study of CT density after
reported to be 2.6 and 1.9% in the concurrent and RT to the thorax for lymphoma and breast cancer
sequential treatment arms, respectively (P = 0.86) found that smokers had significantly smaller changes
(Furuse et al. 1999). It does not appear that the current (P = 0.002); however, there was no significant ven-
standard of platinum-based concurrent chemotherapy tilation or perfusion differences (Theuws et al. 1998).
increases the risk for RT-induced lung injury while A multivariate analysis evaluated SPECT-generated
616 L. Xie et al.

dose response curves and found an increase in control arm were found to have fibrosis on a chest CT
radiation sensitivity in the dose range [40 Gy for scan (53 vs. 28%, P \ 0.005). Equally by important,
non-smokers versus smokers (Garipagaoglu et al. there was no difference noted in tumor response
1999). There has been some speculation that this (Antonadou et al. 2001).
protective effect may be due to a cytokine effect. A separate trial at M.D. Anderson Cancer Center
These observations are not a reason for patients to randomized patients receiving concurrent chemo-
continue smoking while undergoing TRT, however, therapy and hyperfractionated TRT to amifostine or
one day they could help lead to related pharmaco- no amifostine (Komaki et al. 2002). In this trial, acute
logical interventions. pneumonitis was significantly reduced in patients
While many of the methods presented have some treated with amifostine (31 vs. 7.4%, P = 0.03).
utility in predicting RT-induced lung injury, it is There was no difference in the median survival times
likely that a combination of data from several dif- (Komaki et al. 2002).
ferent clinical, biological, and dosimetric functions The radiation therapy oncology group (RTOG
will ultimately provide the most valuable risk 98-01) has recently completed a randomized trial
assessment. For example, as the cytokine cascade in looking at the addition of amifostine to induction
the pathogenesis of RT-induced lung injury becomes carboplatin and paclitaxel (C/P) followed by concur-
better understood, biologic data will be combined rent hyperfractionated TRT and C/P with or without
with dosimetric information and patient-specific lung amifostine. In contrast to earlier studies, this trial
function, which hopefully will lead to improved demonstrated no difference in pneumonitis rates with
prognostication of RT-induced lung injury. or without amifostine Werner-Wasik et al. (2003).
However, in this trial, patients received twice-a-day
TRT, but amifostine only once-a-day, resulting in
4 Modifiers of RT-Induced Lung potential protection for just half of the treatments.
Injury There was also a high drop-out rate of patients in the
treatment arm and when the results were analyzed by
A variety of strategies have been attempted to intent to treat, a large portion did not receive ami-
decrease RT-induced lung toxicity, including dosi- fostine. Because of these caveats, a new trial with
metric variations, pharmacologic agents, and altering once-a-day radiation and subcutaneous amifostine, to
dose using patient-specific biological information. hopefully decrease toxicity, is being initiated. At
There have been several randomized trials of the cy- present, it is unclear if the use of amifostine in
toprotector amifostine (WR-2721) in patients receiv- patients treated with TRT will be of significant benefit
ing TRT. Amifostine (WR-2721) is a phosphorylated in reducing RT-induced lung injury.
aminothiol that demonstrates cytoprotection of normal Assessing changes in biological markers during the
tissues when combined with RT (Wasserman 1999). course of TRT and changing treatment plans
Cytoprotection is believed to result from elimination according to risk categories could potentially lead to a
of free radicals produced by the interaction of ionizing decrease in RT-induced lung toxicity. This has been
RT and water molecules (Capizzi 1999). There is undertaken and a series of patients were treated with
conflicting evidence that amifostine can offer a pneu- twice-daily TRT and based on TGF-b levels during
moprotective benefit in patients receiving TRT. A the course of therapy, RT dose was escalation
randomized trial was performed in Greece using (Anscher et al. 2001). Fourteen patients whose TGF-b
patients with advanced stage lung cancer and who levels were normal after 73.6 Gy were escalated to
received TRT with or without amifostine (Antonadou 80 Gy (n = 8) and 86.4 Gy (n = 6). Overall, the rate
et al. 2001). During the first month after TRT, dyspnea of significant lung toxicity was low in patients with
with minimal exertion was observed in 27% of control stable or declining TGF-b levels, indicating that there
and only 12% of the patients treated with amifostine is the potential to individualize TRT according to
(P = 0.058). After three months, the incidence patient-specific biological factors.
of [ grade 2 pneumonitis was 52% in the control arm In general, patients with lung cancer are simulated
compared to 12% in the amifostine arm (P \ 0.001). for treatment using chest CT scans and standard 3D
At six months, significantly more patients in the planning systems. Fields are arranged to treat the
Radiation-Induced Lung and Heart Toxicity 617

tumor, involving lymph nodes, elective mediastinal, TRT with the incidence of pneumonitis (Willner et al.
and sometimes supraclavicular lymph nodes. 2003). When each lung was analyzed separately, the
Variations of these methods could lead to a decrease in incidence of pneumonitis was highly correlated to the
RT-induced lung injury. For example, limiting volume of ipsilateral lung receiving [40 Gy. In
radiation to only areas with known tumor would contrast, the incidence of pneumonitis decreased as
exclude elective nodal treatment and potentially spare the volume of lung receiving less than 10 Gy
normal lung tissue (Rosenzweig et al. 2001). Treating increased. These results indicate that it is reasonable
only positron emission tomography (PET), positive to spread low doses of RT outside the target area and
nodal disease has been attempted with no apparent in this study, it appeared that reducing the volume of
change in tumor control and low pneumonitis rates lung receiving [40 Gy and increasing the volume
(Belderbos et al. 2003). Limiting elective nodal receiving \10 Gy will lead to less RT-induced lung
irradiation is a simple method to decrease the potential injury (Willner et al. 2003). These data could be the
for RT-induced lung injury and should become a more basis for DVH constraints in IMRT (Willner et al.
widespread technique for patients receiving TRT 2003). A separate study compared dose escalation
(De Ruysscher et al. 2005; van Loon et al. 2010). strategies using either 3D treatment planning or
A method to use the information from ventilation/ IMRT using the same dose constraint of
perfusion scans to decrease dose to the most func- MLD \24 Gy (Marnitz et al. 2002). It was possible to
tional portion of the lung has the potential to reduce give higher doses to the target volume while keeping
RT-induced lung injury. While most treatment plans within the MLD restriction using IMRT (Marnitz
are developed with this intention, it is difficult to et al. 2002). A similar study compared IMRT to 3D
accomplish with the present day technology. Perfu- treatment planning as well as to traditional treatment
sion-weighted optimization using perfusion dose- planning with elective nodal irradiation (Grills et al.
functional histograms (DFHs) has been attempted and 2003). When meeting all of the standard normal-tis-
the results appear promising (Seppenwoolde et al. sue constraints, IMRT delivered 25–35% higher dose
2002). Thus, as more sophisticated treatment planning to the target compared to 3D and [100% higher than
systems are developed, better tailoring of dose using standard treatment planning including elective nodal
radiological/physiologic data may reduce RT-induce irradiation (Grills et al. 2003). In the near future it is
lung injury. Intensity modulated radiation therapy likely that a significant amount of prospective data
(IMRT) has been suggested as a means to steer inci- will be available regarding the benefit (or lack of)
dental dose away from better-functioning regions of IMRT planning to reduce RT-induced lung injury.
the lung (Yin et al. 2010; Das et al. 2004). Two relatively recent non-randomized studies have
Several methods have been attempted to eliminate suggested lower rates of Radiation Pneumonitis with
the need for larger treatment volumes to compensate IMRT versus conventional 3D conformal approaches
for respiratory motion. Respiratory gating is the tim- (Yom et al. 2007; Liao et al. 2010).
ing of TRT with the respiratory cycle and the deep
inspiration breath-hold technique maintains the GTV
in the same position during treatment (Ford et al. 5 Cardiotoxic Effects of Thoracic
2002; Yorke et al. 2002). Gating, possibly together Radiation Therapy
with IMRT, may allow for a more conformal treat-
ment plan and, thus, reduce the potential risks of Heart injury is an inherent risk in the treatment of
treatment. Further evaluations of these techniques are lung cancer arising from the use of TRT, either alone
necessary before they become widely accepted. or in combination with cardiotoxic chemotherapeutic
The use of IMRT is becoming more widespread agents. At least a portion of the heart is typically
and it has been used in TRT. However, the long- exposed to a relatively high dose of radiation when
standing question of whether a little dose to a large the mediastinum and/or primary lung tumors are tar-
volume of normal lung is better than a high dose to a geted. However, cardiac injury is not commonly
smaller volume of lung has not been answered. reported in patients who receive TRT for lung cancer.
An analysis of this issue was recently performed by There are two primary reasons for this. First, most
comparing varying doses to the normal lung during patients treated with TRT for unresectable lung
618 L. Xie et al.

cancer have a short life expectancy. Second, patients controls (Boivin and Hutchison 1982; Boivin et al.
treated for lung cancer typically have pre-existing 1992; Brierley et al. 1998; Gustavsson et al. 1990;
cardiopulmonary disease, and subsequent functional Hancock et al. 1993a, 1993b; Hancock and Hoppe
deterioration is typically ascribed to pre-existing 1996; Henry-Amar et al. 1990; Ng et al. 2002; Po-
disease, RT-induced lung dysfunction, and/or tumor hjola-Sintonen et al. 1987). The relative risk of death
progression, rather than to cardiotoxicity. As our from AMI for patients treated as children was even
ability to successfully treat lung cancer and concur- higher (41.5), reflecting the increased sensitivity of
rent pulmonary disease/injury improves, minimizing children to the cardiotoxic effects of radiation and/or
cardiotoxicity will become an important goal of the the low baseline risk of AMI in the general population
thoracic radiation oncologist. below age 50 (Hancock et al. 1993) (Aleman et al.
Cardiac injury in irradiated lung cancer patients 2007; Swerdlow et al. 2007). Pericarditis has also
has not been well studied for the reasons described been reported following mediastinal irradiation for
above. However, the late effects of radiotherapy on Hodgkin’s disease, and its incidence is strongly
the heart have been extensively studied in survivors of related to the volume of heart irradiated (Carmel and
Hodgkin’s disease and breast cancer. While the Kaplan 1976). The results of these studies have had a
radiotherapy fields and doses used in the treatment of significant impact on the management of Hodgkin’s
Hodgkin’s disease and breast cancer differ markedly disease today. Many of the patients in these studies
from those used to treat lung cancer, these studies were treated with radiotherapy alone to doses in
illustrate the fundamental principles of radiation- excess of 40 Gy. The current treatment approaches
induced cardiotoxicity which may be applicable to emphasize combination chemotherapy followed by
patients with lung cancer. low-dose consolidative RT, in part to reduce the
Incidental cardiac irradiation has been strongly expected long-term cardiac toxicity of treatment.
associated with the development of pericarditis and An increased risk of cardiac death, in particular
premature coronary artery disease, and weak associ- AMI, has also been seen in older trials of post-mas-
ations also exist for a wide range of clinical syn- tectomy RT, particularly for left-sided breast cancer
dromes including cardiomyopathy, valvular disease, (Gyenes 1998; Cuzick et al. 1987, 1994; Rutqvist and
conduction system abnormalities, and autonomic Johansson 1990; Rutqvist et al. 1992; Early Breast
dysfunction. When they occur following the treatment Cancer Trialists’ Collaborative Group 1990, 1995,
of children or adolescents, these syndromes are often 2000; Host et al. 1986; Jones and Ribeiro 1989;
distinguished by their early age of onset. When older Paszat et al. 1998). These older trials used RT tech-
patients are irradiated, the resultant cardiac syn- niques that resulted in a larger volume of heart in the
dromes are generally indistinguishable on clinical RT field than is typically seen with modern treatment
grounds from the more usual forms of the disease. approaches. As a result, reductions in breast cancer
Although changes in the structure and function of the deaths in these trials were offset by increases in car-
intrathoracic viscera after TRT should be considered, diac deaths, such that post-mastectomy RT had a
the manifestations of TRT-induced heart disease are detrimental effect on the overall survival. With more
essentially treated the same as the more usual forms modern RT techniques, cardiac toxicity appears to
of heart disease (Adams et al. 2003a, 2003b). have been reduced, resulting in a net mortality benefit
An increased risk of death from acute myocardial following post-mastectomy RT (Overgaard et al.
infarction (AMI) has been observed in long-term 1997, 1999; Ragaz et al. 1997; Whelan et al. 2000).
survivors of Hodgkin’s disease treated with radio- With modern RT approaches, however, cardiac tox-
therapy fields that encompassed at least part of the icity may result. Modern techniques typically incor-
heart. Mediastinal radiation fields typically used for porate tangential fields that incidentally include the
Hodgkin’s disease deliver 20–40 Gy to the medial anterior myocardium. Furthermore, some patients are
aspect of the heart, and thus include the ostium of the treated with beams that are directed from the anterior
coronary arteries. Occasionally, the remainder of the direction towards the medial breast/chest-wall and
heart may receive a lesser dose. In patients treated as internal mammary lymph nodes. Typical doses are
adults, the relative risk for death from AMI ranged 45–50 Gy. The incidence of cardiac dysfunction fol-
2.6–14.9 compared with age and gender-matched lowing radiation for breast cancer is related to the
Radiation-Induced Lung and Heart Toxicity 619

volume of heart irradiated (Rutqvist et al. 1992; Gyenes of radiation-associated cardiac events in these patients.
et al. 1998). Pericarditis has also been reported in Nevertheless, there are some data that demonstrate that
these patients. this may be an important clinical problem. In a meta-
Subclinical cardiac injury is very common. Non- analysis, post-operative RT was associated with a 6%
lethal symptomatic cardiac injury is reported to occur increased rate of mortality, cause not specified (PORT
in 0–50% of patients receiving incidental cardiac Meta-analysis Trialists Group 1998). In a randomized
irradiation during treatment for Hodgkin’s disease, clinical trial assessing the utility of post-operative
carcinoma of the breast, lung, esophagus, or medul- TRT, the addition of RT increased the rate of cardiac
loblastoma (Pohjola-Sintonen et al. 1987; Carmel and mortality three fold compared to non-irradiated con-
Kaplan 1976; Cosset et al. 1988, 1991; Applefeld and trols. Five percent of the irradiated patients died of
Wiernik 1983; Jakacki et al. 1993; Yu et al. 2003). cardiac disease; non-lethal morbidity was not addres-
Among asymptomatic patients, subclinical damage sed (Dautzenberg et al. 1999). Cardiac toxicity has not
can be detected by electrocardiogram (EKG), echo- been reported in patients treated definitively for lung
cardiogram (ECHO), or other radiological studies in cancer, but has been reported in the post-operative
approximately 0–67% (Carmel and Kaplan 1976; setting where survival rates are higher (Dautzenberg
Strender et al. 1986; Constine et al. 1997; Gomez et al. 1999). This observation supports the concept that
et al. 1983; Gottdiener et al. 1983; Lagrange et al. such cardiac events may be under-reported in long-
1992; van Rijswijk et al. 1987; Hardenbergh et al. term survivors of lung cancer.
2001; Makinen et al. 1990; Watchie et al. 1987). The concern for RT-induced cardiotoxicity is
We and others have used SPECT cardiac perfusion heightened by the widespread use of potentially car-
imaging as a means to detect microvascular injury in diotoxic systemic therapy and the high prevalence of
the myocardium. In patients irradiated for left-sided cardiac risk factors in the lung cancer patient popu-
breast cancer, approximately 50–75% of patients will lation. Paclitaxel, a widely used agent in the treatment
develop new perfusion defects if C5% of the left of lung cancer, is potentially cardiotoxic (Vogt et al.
ventricle is included within the radiation field (Marks 1996; Kelly et al. 1997). A variety of clinical factors
et al. 2003). These defects appear to be associated (age, male gender, tobacco use, obesity, diabetes
with the corresponding abnormalities in wall motion mellitus, family history, hypercholesterolemia, and
and the possibly subtle reductions in ejection fraction. hypertension) have been associated with an increased
With longer follow-up, most of these perfusion incidence of ischemic cardiac disease. Many of these
defects persist (Prosnitz et al. 2007). factors such as, hypertension (Lauk and Trott 1988),
The reported incidence of RT-induced cardiac lack of exercise (Geist et al. 1990), and high choles-
toxicity varies widely depending on the endpoint terol/fat diet (Artom et al. 1965; Amromin et al.
used. The reported frequency of cardiac morbidity 1964), may increase the risk of RT-induced cardiac
also depends on the population of patients considered. injury (Hull et al. 2003).
Studies that report on a group of patients seen by A clinical study from Memorial Sloan Kettering
cardiologists tend to overestimate the incidence, since Cancer Center suggests that the dose to the inferior
asymptomatic patients are often not included. Con- lung is more predictive for radiation pneumonitis than
versely, retrospective studies of patients evaluated is dose delivered to the superior aspect of the lung
some years following RT tend to underestimate the (Yorke et al. 2002). A similar finding has been
incidence since only the evaluable survivors are reported in mice (Tucker et al. 1997; Travis et al.
included. Nevertheless, the preponderance of the data 1997). It is possible that irradiation of the inferior-
suggests that RT-induced cardiac damage, either lung may be a barometer for incidental cardiac irra-
clinical or subclinical, is common. diation, as the heart is located in the inferior chest in
The experience with Hodgkin’s disease and breast both mice and humans. Interestingly, in rats (where
cancer demonstrates the potential impact of TRT on the heart is located in the superior left hemi-thorax),
cardiac function in patients irradiated for lung cancer. irradiation of the superior or left lung resulted in more
As outlined above, the generally poor survival rates ‘‘lung’’ toxicity than did similar treatment to the right
and concurrent illnesses in patients irradiated for lung or inferior lung (Jiresova et al. 2002). In concert,
cancer probably account for the low reported incidence these data suggest that incidental cardiac irradiation
620 L. Xie et al.

competing morbidity/mortality. The possibility of


RT-associated cardiac dysfunction should be consid-
ered in patients who have been irradiated for lung
cancer. Additional study is needed to better understand
the clinical importance of such injury.

Acknowledgment This work was supported by NIH Grant


2R201 CA69579-09.

References

Abid SH, Malhotra V, Perry MC (2001) Radiation-induced and


chemotherapy-induced pulmonary injury. Curr Opin Oncol
13:242–248
Abratt RP, Willcox PA, Smith JA (1990) Lung cancer in
patients with borderline lung functions–zonal lung perfusion
scans at presentation and lung function after high dose
irradiation. Radiother Oncol 19:317–322
Adams MJ, Lipshultz SE, Schwartz C et al (2003a) Radiation-
associated cardiovascular disease: manifestations and man-
agement. Semin Radiat Oncol 13:346–356
Fig. 1 Non-axial beams to limit cardiac dose Adams MJ, Hardenbergh PH, Constine LS et al (2003b)
Radiation-associated cardiovascular disease. Crit Rev Oncol
Hematol 45:55–75
may result in subclinical injury that masquerades as, Aleman BM, van den Belt-Dusebout AW, De Bruin ML et al
or interacts with, ‘‘lung’’ toxicity. (2007) Late cardiotoxicity after treatment for Hodgkin
In light of these issues, we recommend that care be lymphoma. Blood 109:1878–1886
taken to minimize incidental cardiac irradiation dur- Amromin GD, Gildenhorn HL, Solomon RD et al (1964) The
synergism of X-irradiation and cholesterol-fat feeding on
ing TRT for lung cancer. Towards this goal, we often the development of coronary artery lesions. J Atheroscler
use non-axial beams to minimize incidental cardiac Res 24:325–334
irradiation. This is most useful in patients with lower Anscher MS, Murase T, Prescott DM et al (1994) Changes in
lobe tumors with hilar/mediastinal nodes. In this set- plasma TGF beta levels during pulmonary radiotherapy as a
predictor of the risk of developing radiation pneumonitis.
ting, one is often able to shadow the primary tumor Int J Radiat Oncol Biol Phys 30:671–676
with the nodal disease and therefore the beam aper- Anscher MS, Marks LB, Shafman TD et al (2001) Using
ture can actually be smaller with non-axial beams plasma transforming growth factor beta-1 during radiother-
than with axial beams. For example, a primary tumor apy to select patients for dose escalation. J Clin Oncol
19:3758–3765
in the left lower lobe with metastases to the left hilum Antonadou D, Coliarakis N, Synodinou M et al (2001)
and pre-carinal area, can often be treated with oblique Randomized phase III trial of radiation treatment ± ami-
fields oriented from right-anterior–superior and fostine in patients with advanced-stage lung cancer. Int J
opposed left-posterior-inferior directions, resulting in Radiat Oncol Biol Phys 51:915–922
Applefeld MM, Wiernik PH (1983) Cardiac disease after
irradiation of a smaller cardiac volume (Fig. 1). radiation therapy for Hodgkin’s disease: analysis of 48
It is important to remember that incidental cardiac patients. Am J Cardiol 51:1679–1681
irradiation is a concern for tumors in both the left and Artom C, Lofland HB Jr, Clarkson TB (1965) Ionizing
right thorax. Off-cord oblique axial boost fields for radiation, atherosclerosis, and lipid metabolism in pigeons.
Radiat Res 26:165–177
right lung tumors usually include the anterior heart.
Barriger RB, Fakiris AJ, Hanna N et al (2010) Dose–Volume
Given the degree of cardiac injury observed in Analysis of Radiation Pneumonitis in Non–Small-Cell Lung
patients irradiated for breast cancer, Hodgkin’s disease Cancer Patients Treated With Concurrent Cisplatinum and
and other mediastinal neoplasms, it is extremely likely Etoposide With or Without Consolidation Docetaxel. Inter-
national Journal of Radiation OncologyBiologyPhysics
that similar events are occurring in patients irradiated
78:1381–1386
for lung cancer. To date, this has not been recognized Belderbos JSA, De Jaeger K, Heemsbergen WD et al (2003)
as an important clinical problem, due primarily to First results of a phase I/II dose escalation trial in non-small
Radiation-Induced Lung and Heart Toxicity 621

cell lung cancer using three-dimensional conformal radio- participated in trials of radiotherapy. J Clin Oncol
therapy. Radiother Oncol 66:119–126 12:447–453
Bell J, McGivern D, Bullimore J et al (1988) Diagnostic Das SK, Bell M, Marks LB et al (2004) A preliminary study of
imaging of post-irradiation changes in the chest. Clin Radiol the role of modulated electron beams in intensity modulated
39:109–119 radiotherapy, using automated beam orientation and modal-
Boivin JF, Hutchison GB (1982) Coronary heart disease ity selection. Int J Radiat Oncol Biol Phys 59:602–617
mortality after irradiation for Hodgkin’s disease. Cancer Dautzenberg B, Arriagada R, Chammard AB et al (1999) A
49:2470–2475 controlled study of postoperative radiotherapy for patients
Boivin JF, Hutchison GB, Lubin JH et al (1992) Coronary with completely resected nonsmall cell lung carcinoma.
artery disease mortality in patients treated for Hodgkin’s Groupe d’Etude et de Traitement des Cancers Bronchiques.
disease. Cancer 69:1241–1247 Cancer 86:265–273
Bonnet RB, Bush D, Cheek GA et al (2001) Effects of proton De Ruysscher D, Wanders S, van Haren E et al (2005) Selective
and combined proton/photon beam radiation on pulmonary mediastinal node irradiation based on FDG-PET scan data
function in patients with resectable but medically inoperable in patients with non-small-cell lung cancer: a prospective
non-small cell lung cancer. Chest 120:1803–1810 clinical study. Int J Radiat Oncol Biol Phys 62:988–994
Brach MA, Hass R, Sherman ML et al (1991) Ionizing radiation Dechambre St, Dorzee J, Fastrez J et al (1998) Bronchial
induces expression and binding activity of the nuclear factor stenosis and sclerosing mediastinitis: an uncommon com-
kappa B. J Clin Invest 88:691–695 plication of external thoracic radiotherapy. Eur Respir J
Brady LW, Germon PA, Cander L (1965) The Effects of 11:1188–1190
Radiation Therapy on Pulmonary Function in Carcinoma of Dillman RO, Seagren SL, Propert KJ et al (1990) A randomized
the Lung. Radiology 85:130–134 trial of induction chemotherapy plus high-dose radiation
Brierley JD, Rathmell AJ, Gospodarowicz MK et al (1998) Late versus radiation alone in stage III non-small-cell lung
effects of treatment for early-stage Hodgkin’s disease. Br J cancer. N Engl J Med 323:940–945
Cancer 77:1300–1310 Early Breast Cancer Trialists’ Collaborative Group (1990)
Byhardt RW, Martin L, Pajak TF et al (1993) The influence of Treatment of early breast cancer. vol 1. Oxford University,
field size and other treatment factors on pulmonary toxicity UK
following hyperfractionated irradiation for inoperable non- Early Breast Cancer Trialists’ Collaborative Group (1995)
small cell lung cancer (NSCLC)–analysis of a Radiation Effects of radiotherapy and surgery in early breast cancer.
Therapy Oncology Group (RTOG) protocol. Int J Radiat An overview of the randomized trials. N Engl J Med 333:
Oncol Biol Phys 27:537–544 1444-1455
Capizzi RL (1999) Amifostine reduces the incidence of Early Breast Cancer Trialists’ Collaborative Group (2000)
cumulative nephrotoxicity from cisplatin: laboratory and Favourable and unfavourable effects on long-term survival
clinical aspects. Semin Oncol 26:72–81 of radiotherapy for early breast cancer: an overview of the
Carmel RJ, Kaplan HS (1976) Mantle irradiation in Hodgkin’s randomised trials. Lancet 355:1757-1770
disease. An analysis of technique, tumor eradication, and Epperly MW, Travis EL, Sikora C et al (1999) Manganese
complications. Cancer 37:2813–2825 [correction of Magnesium] superoxide dismutase
Chen Y, Williams J, Ding I et al (2002) Radiation pneumonitis (MnSOD) plasmid/liposome pulmonary radioprotective
and early circulatory cytokine markers. Semin Radiat Oncol gene therapy: modulation of irradiation-induced mRNA
12:26–33 for IL-I, TNF-alpha, and TGF-beta correlates with delay of
Choi NC, Kanarek DJ (1994) Toxicity of thoracic radiotherapy organizing alveolitis/fibrosis. Biol Blood Marrow Trans-
on pulmonary function in lung cancer. Lung Cancer plant 5:204–214
10(Suppl 1):219–230 Evans ES, Kocak Z, Zhou S-M et al (2006) Does transforming
Constine LS, Schwartz RG, Savage DE et al (1997) Cardiac growth factor-beta1 predict for radiation-induced pneumo-
function, perfusion, and morbidity in irradiated long-term nitis in patients treated for lung cancer? Cytokine 35:
survivors of Hodgkin’s disease. Int J Radiat Oncol Biol 186–192
Phys 39:897–906 Fan M, Marks LB, Hollis D et al (2001) Can we predict
Cosset JM, Henry-Amar M, Girinski T et al (1988) Late radiation-induced changes in pulmonary function based on
toxicity of radiotherapy in Hodgkin’s disease. The role of the sum of predicted regional dysfunction? J Clin Oncol
fraction size. Acta Oncol 27:123–129 19:543–550
Cosset JM, Henry-Amar M, Pellae-Cosset B et al (1991) Favaretto A, Paccagnella A, Tomio L et al (1996) Pre-operative
Pericarditis and myocardial infarctions after Hodgkin’s chemoradiotherapy in non-small cell lung cancer stage III
disease therapy. Int J Radiat Oncol Biol Phys 21:447–449 patients. Feasibility, toxicity and long-term results of a
Curran WJ, Moldofsky PJ, Solin LJ (1992) Observations on the phase II study. Eur J Cancer 32A:2064–2069
predictive value of perfusion lung scans on post-irradiation Finkelstein JN, Johnston CJ, Baggs R et al (1994) Early
pulmonary function among 210 patients with bronchogenic alterations in extracellular matrix and transforming growth
carcinoma. Int J Radiat Oncol Biol Phys 24:31–36 factor beta gene expression in mouse lung indicative of late
Cuzick J, Stewart H, Peto R et al (1987) Overview of radiation fibrosis. Int J Radiat Oncol Biol Phys 28:621–631
randomized trials of postoperative adjuvant radiotherapy Ford EC, Mageras GS, Yorke E et al (2002) Evaluation of
in breast cancer. Cancer Treat Rep 71:15–29 respiratory movement during gated radiotherapy using film
Cuzick J, Stewart H, Rutqvist L et al (1994) Cause-specific and electronic portal imaging. Int J Radiat Oncol Biol Phys
mortality in long-term survivors of breast cancer who 52:522–531
622 L. Xie et al.

Franko AJ, Sharplin J, Ghahary A et al (1997) Immunohisto- to ionizing radiation. Proc Natl Acad Sci U S A
chemical localization of transforming growth factor beta 86:10104–10107
and tumor necrosis factor alpha in the lungs of fibrosis- Hallahan DE, Virudachalam S, Beckett M et al (1991)
prone and ‘‘non-fibrosing’’ mice during the latent period and Mechanisms of X-ray-mediated protooncogene c-jun
early phase after irradiation. Radiat Res 147:245–256 expression in radiation-induced human sarcoma cell lines.
Fu XL, Jiang GL, Wang LJ et al (1997) Hyperfractionated Int J Radiat Oncol Biol Phys 21:1677–1681
accelerated radiation therapy for non-small cell lung cancer: Hallahan DE, Virudachalam S, Kufe DW et al (1994)
clinical phase I/II trial. Int J Radiat Oncol Biol Phys Ketoconazole attenuates radiation-induction of tumor
39:545–552 necrosis factor. Int J Radiat Oncol Biol Phys 29:777–780
Fu XL, Huang H, Bentel G et al (2001) Predicting the risk of Hancock SL, Hoppe RT (1996) Long-Term Complications of
symptomatic radiation-induced lung injury using both the Treatment and Causes of Mortality After Hodgkin’s
physical and biologic parameters V(30) and transforming Disease. Semin Radiat Oncol 6:225–242
growth factor beta. Int J Radiat Oncol Biol Phys 50:899–908 Hancock SL, Donaldson SS, Hoppe RT (1993a) Cardiac
Furuse K, Fukuoka M, Kawahara M et al (1999) Phase III study disease following treatment of Hodgkin’s disease in chil-
of concurrent versus sequential thoracic radiotherapy in dren and adolescents. J Clin Oncol 11:1208–1215
combination with mitomycin, vindesine, and cisplatin in Hancock SL, Tucker MA, Hoppe RT (1993b) Factors affecting
unresectable stage III non-small-cell lung cancer. J Clin late mortality from heart disease after treatment of Hodg-
Oncol 17:2692–2699 kin’s disease. Jama 270:1949–1955
Garipagaoglu M, Munley MT, Hollis D et al (1999) The effect Hardenbergh PH, Munley MT, Bentel GC et al (2001) Cardiac
of patient-specific factors on radiation-induced regional perfusion changes in patients treated for breast cancer with
lung injury. Int J Radiat Oncol Biol Phys 45:331–338 radiation therapy and doxorubicin: preliminary results. Int J
Geist BJ, Lauk S, Bornhausen M et al (1990) Physiologic Radiat Oncol Biol Phys 49:1023–1028
consequences of local heart irradiation in rats. Int J Radiat Haston CK, Zhou X, Gumbiner-Russo L et al (2002) Universal
Oncol Biol Phys 18:1107–1113 and radiation-specific loci influence murine susceptibility to
Gomez GA, Park JJ, Panahon AM et al (1983) Heart size and radiation-induced pulmonary fibrosis. Cancer Res
function after radiation therapy to the mediastinum in patients 62:3782–3788
with Hodgkin’s disease. Cancer Treat Rep 67:1099–1103 Henry-Amar M, Hayat M, Meerwaldt JH et al (1990) Causes of
Gottdiener JS, Katin MJ, Borer JS et al (1983) Late cardiac death after therapy for early stage Hodgkin’s disease
effects of therapeutic mediastinal irradiation. Assessment by entered on EORTC protocols. EORTC Lymphoma Coop-
echocardiography and radionuclide angiography. N Engl J erative Group. Int J Radiat Oncol Biol Phys 19:1155–1157
Med 308:569–572 Hernando ML, Marks LB, Bentel GC et al (2001) Radiation-
Graham MV, Purdy JA, Emami B et al (1999) Clinical dose- induced pulmonary toxicity: a dose-volume histogram
volume histogram analysis for pneumonitis after 3D treat- analysis in 201 patients with lung cancer. Int J Radiat
ment for non-small cell lung cancer (NSCLC). Int J Radiat Oncol Biol Phys 51:650–659
Oncol Biol Phys 45:323–329 Hong JH, Chiang CS, Campbell IL et al (1995) Induction of
Graves PR, Siddiqui F, Anscher MS et al (2010) Radiation acute phase gene expression by brain irradiation. Int J
Pulmonary Toxicity: from Mechanisms to Management. Radiat Oncol Biol Phys 33:619–626
Semin Radiat Oncol 20:201–207 Hong JH, Chiang CS, Sun JR et al (1997) Induction of c-fos and
Grills IS, Yan D, Martinez AA et al (2003) Potential for junB mRNA following in vivo brain irradiation. Brain Res
reduced toxicity and dose escalation in the treatment of Mol Brain Res 48:223–228
inoperable non-small-cell lung cancer: a comparison of Host H, Brennhovd IO, Loeb M (1986) Postoperative radio-
intensity-modulated radiation therapy (IMRT), 3D confor- therapy in breast cancer–long-term results from the Oslo
mal radiation, and elective nodal irradiation. Int J Radiat study. Int J Radiat Oncol Biol Phys 12:727–732
Oncol Biol Phys 57:875–890 Hull MC, Morris CG, Pepine CJ et al (2003) Valvular
Gross NJ (1977) Pulmonary effects of radiation therapy. Ann dysfunction and carotid, subclavian, and coronary artery
Intern Med 86:81–92 disease in survivors of hodgkin lymphoma treated with
Gross NJ (1980) Experimental radiation pneumonitis. IV. radiation therapy. Jama 290:2831–2837
Leakage of circulatory proteins onto the alveolar surface. Inoue A, Kunitoh H, Sekine I et al (2001) Radiation pneumo-
J Lab Clin Med 95:19–31 nitis in lung cancer patients: a retrospective study of risk
Gustavsson A, Eskilsson J, Landberg T et al (1990) Late factors and the long-term prognosis. Int J Radiat Oncol Biol
cardiac effects after mantle radiotherapy in patients with Phys 49:649–655
Hodgkin’s disease. Ann Oncol 1:355–363 Jakacki RI, Goldwein JW, Larsen RL et al (1993) Cardiac
Gyenes G (1998) Radiation-induced ischemic heart disease in dysfunction following spinal irradiation during childhood.
breast cancer–a review. Acta Oncol 37:241–246 J Clin Oncol 11:1033–1038
Gyenes G, Rutqvist LE, Liedberg A et al (1998) Long-term Jin H, Tucker SL, Liu HH et al (2009) Dose–volume thresholds
cardiac morbidity and mortality in a randomized trial of pre- and smoking status for the risk of treatment-related pneu-
and postoperative radiation therapy versus surgery alone in monitis in inoperable non-small cell lung cancer treated with
primary breast cancer. Radiother Oncol 48:185–190 definitive radiotherapy. Radiother Oncol 91:427–432
Hallahan DE, Spriggs DR, Beckett MA et al (1989) Increased Jiresova A, Wiegman EM, Kampinga HH et al (2002) Dose-
tumor necrosis factor alpha mRNA after cellular exposure volume-region effects in partial irradiation of the rat lung.
Radiation-Induced Lung and Heart Toxicity 623

Programs Abstr Radiat Res Soc North Am Hyperth Soc Makinen L, Makipernaa A, Rautonen J et al (1990) Long-term
103:114 cardiac sequelae after treatment of malignant tumors with
Johansson S, Bjermer L, Franzen L et al (1998) Effects of radiotherapy or cytostatics in childhood. Cancer
ongoing smoking on the development of radiation-induced 65:1913–1917
pneumonitis in breast cancer and oesophagus cancer Marks LB, Spencer DP, Bentel GC et al (1993) The utility of
patients. Radiother Oncol 49:41–47 SPECT lung perfusion scans in minimizing and assessing
Johnston CJ, Williams JP, Okunieff P et al (2002) Radiation- the physiologic consequences of thoracic irradiation. Int J
induced pulmonary fibrosis: examination of chemokine and Radiat Oncol Biol Phys 26:659–668
chemokine receptor families. Radiat Res 157:256–265 Marks LB, Hollis D, Munley M et al (2000) The role of lung
Jones JM, Ribeiro GG (1989) Mortality patterns over 34 years perfusion imaging in predicting the direction of radiation-
of breast cancer patients in a clinical trial of post-operative induced changes in pulmonary function tests. Cancer
radiotherapy. Clin Radiol 40:204–208 88:2135–2141
Katzenstein A-LA, Askin FB (2006) Katzenstein and Askin’s Marks LB, Yu X, Zhou S (2003) The impact of irradiated left
surgical pathology of non-neoplastic lung disease, 4th edn. ventricular volume on the incidence of radiation-induced
Elsevier Saunders, Philadelphia cardiac perfusion changes. Int J Radiat Oncol Biol Phys
Kelly K, Pan Z, Murphy J et al (1997) A phase I trial of 57:S129
paclitaxel plus carboplatin in untreated patients with Marks LB, Bentzen SM, Deasy JO et al (2010) Radiation dose-
advanced non-small cell lung cancer. Clin Cancer Res volume effects in the lung. Int J Radiat Oncol Biol Phys
3:1117–1123 76:S70–S76
Komaki R, Lee JS, Kaplan B et al (2002) Randomized phase III Marnitz S, Stuschke M, Bohsung J et al (2002) Intraindividual
study of chemoradiation with or without amifostine for comparison of conventional three-dimensional radiotherapy
patients with favorable performance status inoperable stage and intensity modulated radiotherapy in the therapy of
II-III non-small cell lung cancer: preliminary results. Semin locally advanced non-small cell lung cancer a planning
Radiat Oncol 12:46–49 study. Strahlenther Onkol 178:651–658
Kwon HC, Kim SK, Chung WK et al (2000) Effect of Martel MK, Ten Haken RK, Hazuka MB et al (1994) Dose-
pentoxifylline on radiation response of non-small cell lung volume histogram and 3D treatment planning evaluation of
cancer: a phase III randomized multicenter trial. Radiother patients with pneumonitis. Int J Radiat Oncol Biol Phys
Oncol 56:175–179 28:575–581
Lagrange JL, Darcourt J, Benoliel J et al (1992) Acute cardiac Mattson K, Holsti LR, Poppius H et al (1987) Radiation
effects of mediastinal irradiation: assessment by radionu- pneumonitis and fibrosis following split-course radiation
clide angiography. Int J Radiat Oncol Biol Phys 22:897–903 therapy for lung cancer. A radiologic and physiologic study.
Lauk S, Trott KR (1988) Radiation induced heart disease in Acta Oncol 26:193–196
hypertensive rats. Int J Radiat Oncol Biol Phys 14:109–114 McDonald S, Rubin P, Phillips TL et al (1995) Injury to the
Liao ZX, Komaki RR, Thames HD Jr et al (2010) Influence of lung from cancer therapy: clinical syndromes, measurable
technologic advances on outcomes in patients with unre- endpoints, and potential scoring systems. Int J Radiat Oncol
sectable, locally advanced non–small-cell lung cancer Biol Phys 31:1187–1203
receiving concomitant chemoradiotherapy. Int J Radiat Miller KL, Shafman TD, Anscher MS et al (2005) Bronchial
Oncol Bio Phys 76:775–781 stenosis: an underreported complication of high-dose
Libshitz HI, Shuman LS (1984) Radiation-induced pulmonary external beam radiotherapy for lung cancer? Int J Radiat
change: CT findings. J Comput Assist Tomogr 8:15–19 Oncol Biol Phys 61:64–69
Lind PA, Marks LB, Hollis D et al (2002) Receiver operating Monson JM, Stark P, Reilly JJ et al (1998) Clinical radiation
characteristic curves to assess predictors of radiation- pneumonitis and radiographic changes after thoracic radi-
induced symptomatic lung injury. Int J Radiat Oncol Biol ation therapy for lung carcinoma. Cancer 82:842–850
Phys 54:340–347 Morgan GW, Breit SN (1995) Radiation and the lung: a
Maguire PD, Marks LB, Sibley GS et al (2001) 73.6 Gy and reevaluation of the mechanisms mediating pulmonary
beyond: hyperfractionated, accelerated radiotherapy for injury. Int J Radiat Oncol Biol Phys 31:361–369
non-small-cell lung cancer. J Clin Oncol 19:705–711 Ng AK, Bernardo MP, Weller E et al (2002) Long-term
Mah K, Poon PY, Van Dyk J et al (1986) Assessment of acute survival and competing causes of death in patients with
radiation-induced pulmonary changes using computed early-stage Hodgkin’s disease treated at age 50 or younger.
tomography. J Comput Assist Tomogr 10:736–743 J Clin Oncol 20:2101–2108
Mah K, Van Dyk J, Keane T et al (1987) Acute radiation- Nyman J, Bergman B, Mercke C (1998) Accelerated hyper-
induced pulmonary damage: a clinical study on the response fractionated radiotherapy combined with induction and
to fractionated radiation therapy. Int J Radiat Oncol Biol concomitant chemotherapy for inoperable non-small-cell
Phys 13:179–188 lung cancer–impact of total treatment time. Acta Oncol
Mah K, Keane TJ, Van Dyk J et al (1994) Quantitative effect of 37:539–545
combined chemotherapy and fractionated radiotherapy on Oetzel D, Schraube P, Hensley F et al (1995) Estimation of
the incidence of radiation-induced lung damage: a prospec- pneumonitis risk in three-dimensional treatment planning
tive clinical study. Int J Radiat Oncol Biol Phys 28:563–574 using dose-volume histogram analysis. Int J Radiat Oncol
Makimoto T, Tsuchiya S, Hayakawa K et al (1999) Risk factors Biol Phys 33:455–460
for severe radiation pneumonitis in lung cancer. Jpn J Clin Overgaard M, Hansen PS, Overgaard J et al (1997) Postoper-
Oncol 29:192–197 ative radiotherapy in high-risk premenopausal women with
624 L. Xie et al.

breast cancer who receive adjuvant chemotherapy. Danish Rubin P, Johnston CJ, Williams JP et al (1995) A perpetual
Breast Cancer Cooperative Group 82b Trial. N Engl J Med cascade of cytokines postirradiation leads to pulmonary
337:949–955 fibrosis. Int J Radiat Oncol Biol Phys 33:99–109
Overgaard M, Jensen MB, Overgaard J et al (1999) Postoper- Rutqvist LE, Johansson H (1990) Mortality by laterality of
ative radiotherapy in high-risk postmenopausal breast- the primary tumour among 55, 000 breast cancer patients
cancer patients given adjuvant tamoxifen: Danish Breast from the Swedish Cancer Registry. Br J Cancer 61:
Cancer Cooperative Group DBCG 82c randomised trial. 866–868
Lancet 353:1641–1648 Rutqvist LE, Lax I, Fornander T et al (1992) Cardiovascular
Paszat LF, Mackillop WJ, Groome PA et al (1998) Mortality mortality in a randomized trial of adjuvant radiation therapy
from myocardial infarction after adjuvant radiotherapy for versus surgery alone in primary breast cancer. Int J Radiat
breast cancer in the surveillance, epidemiology, and end- Oncol Biol Phys 22:887–896
results cancer registries. J Clin Oncol 16:2625–2631 Segawa Y, Takigawa N, Kataoka M et al (1997) Risk factors
Perez CA, Stanley K, Rubin P et al (1980) A prospective for development of radiation pneumonitis following radia-
randomized study of various irradiation doses and fraction- tion therapy with or without chemotherapy for lung cancer.
ation schedules in the treatment of inoperable non-oat-cell Int J Radiat Oncol Biol Phys 39:91–98
carcinoma of the lung. Preliminary report by the Radiation Seppenwoolde Y, Muller SH, Theuws JC et al (2000) Radiation
Therapy Oncology Group. Cancer 45:2744–2753 dose-effect relations and local recovery in perfusion for
Perry MC, Eaton WL, Propert KJ et al (1987) Chemotherapy patients with non-small-cell lung cancer. Int J Radiat Oncol
with or without radiation therapy in limited small-cell Biol Phys 47:681–690
carcinoma of the lung. N Engl J Med 316:912–918 Seppenwoolde Y, Engelsman M, De Jaeger K et al (2002)
Pohjola-Sintonen S, Totterman KJ, Salmo M et al (1987) Late Optimizing radiation treatment plans for lung cancer using
cardiac effects of mediastinal radiotherapy in patients with lung perfusion information. Radiother Oncol 63:165–177
Hodgkin’s disease. Cancer 60:31–37 Shapiro SJ, Shapiro SD, Mill WB et al (1990) Prospective study
PORT Meta-analysis Trialists Group (1998) Postoperative of long-term pulmonary manifestations of mantle irradia-
radiotherapy in non-small-cell lung cancer: systematic tion. Int J Radiat Oncol Biol Phys 19:707–714
review and meta-analysis of individual patient data from Simpson JR, Francis ME, Perez-Tamayo R et al (1985)
nine randomised controlled trials. Lancet 352:257-263 Palliative radiotherapy for inoperable carcinoma of the
Prato FS, Kurdyak R, Saibil EA et al (1977) Physiological and lung: final report of a RTOG multi-institutional trial. Int J
radiographic assessment during the development of pul- Radiat Oncol Biol Phys 11:751–758
monary radiation fibrosis. Radiology 122:389–397 Strender LE, Lindahl J, Larsson LE (1986) Incidence of heart
Prosnitz RG, Hubbs JL, Evans ES et al (2007) Prospective disease and functional significance of changes in the
assessment of radiotherapy-associated cardiac toxicity in electrocardiogram 10 years after radiotherapy for breast
breast cancer patients: analysis of data 3 to 6 years after cancer. Cancer 57:929–934
treatment. Cancer 110:1840–1850 Sunyach MP, Falchero L, Pommier P et al (2000) Prospective
Ragaz J, Jackson SM, Le N et al (1997) Adjuvant radiotherapy evaluation of early lung toxicity following three-dimen-
and chemotherapy in node-positive premenopausal women sional conformal radiation therapy in non-small-cell lung
with breast cancer. N Engl J Med 337:956–962 cancer: preliminary results. Int J Radiat Oncol Biol Phys
Roach M, Gandara DR, Yuo HS et al (1995) Radiation 48:459–463
pneumonitis following combined modality therapy for lung Swerdlow AJ, Higgins CD, Smith P et al (2007) Myocardial
cancer: analysis of prognostic factors. J Clin Oncol 13: infarction mortality risk after treatment for Hodgkin
2606–2612 disease: a collaborative British cohort study. J Natl Cancer
Robnett TJ, Machtay M, Vines EF et al (2000) Factors Inst 99:206–214
predicting severe radiation pneumonitis in patients receiv- Theuws JC, Kwa SL, Wagenaar AC et al (1998a) Prediction of
ing definitive chemoradiation for lung cancer. Int J Radiat overall pulmonary function loss in relation to the 3D dose
Oncol Biol Phys 48:89–94 distribution for patients with breast cancer and malignant
Rosenzweig KE, Sim SE, Mychalczak B et al (2001) Elective lymphoma. Radiother Oncol 49:233–243
nodal irradiation in the treatment of non-small-cell lung Theuws JC, Kwa SL, Wagenaar AC et al (1998b) Dose-effect
cancer with three-dimensional conformal radiation therapy. relations for early local pulmonary injury after irradiation
Int J Radiat Oncol Biol Phys 50:681–685 for malignant lymphoma and breast cancer. Radiother
Roswit B, White DC (1977) Severe radiation injuries of the Oncol 48:33–43
lung. AJR Am J Roentgenol 129:127–136 Travis EL (1980) The sequence of histological changes in
Rubenstein JH, Richter MP, Moldofsky PJ et al (1988) mouse lungs after single doses of x-rays. Int J Radiat Oncol
Prospective prediction of post-radiation therapy lung func- Biol Phys 6:345–347
tion using quantitative lung scans and pulmonary function Travis EL, Harley RA, Fenn JO et al (1977) Pathologic changes
testing. Int J Radiat Oncol Biol Phys 15:83–87 in the lung following single and multi-fraction irradiation.
Rubin P, Shapiro DL, Finklestein JN et al (1980) The early Int J Radiat Oncol Biol Phys 2:475–490
release of surfactant following lung irradiation of alveolar Travis EL, Liao ZX, Tucker SL (1997) Spatial heterogeneity of
type II cells. Int J Radiat Oncol Biol Phys 6:75–77 the volume effect for radiation pneumonitis in mouse lung.
Rubin P, Siemann DW, Shapiro DL et al (1983) Surfactant Int J Radiat Oncol Biol Phys 38:1045–1054
release as an early measure of radiation pneumonitis. Int J Tsujino K, Hirota S, Endo M et al (2003) Predictive value of
Radiat Oncol Biol Phys 9:1669–1673 dose-volume histogram parameters for predicting radiation
Radiation-Induced Lung and Heart Toxicity 625

pneumonitis after concurrent chemoradiation for lung histogram parameters of the lung in patients with lung
cancer. Int J Radiat Oncol Biol Phys 55:110–115 cancer treated with 3D conformal radiotherapy. Strahlenther
Tucker SL, Liao ZX, Travis EL (1997) Estimation of the spatial Onkol 179:548–556
distribution of target cells for radiation pneumonitis in Woel RT, Munley MT, Hollis D et al (2002) The time course of
mouse lung. Int J Radiat Oncol Biol Phys 38:1055–1066 radiation therapy-induced reductions in regional perfusion:
Tucker SL, Jin H, Wei X et al (2010) Impact of Toxicity Grade a prospective study with [5 years of follow-up. Int J Radiat
and Scoring System on the Relationship Between Mean Oncol Biol Phys 52:58–67
Lung Dose and Risk of Radiation Pneumonitis in a Large Yamada M, Kudoh S, Hirata K et al (1998) Risk factors of
Cohort of Patients With Non–Small Cell Lung Cancer. Int J pneumonitis following chemoradiotherapy for lung cancer.
Radiat Oncol Bio Phys 77:691–698 Eur J Cancer 34:71–75
Van den Brande P, De Ruysscher D, Vansteenkiste J et al Yin L, Shcherbinin S, Celler A et al (2010) Incorporating
(1998) Sequential treatment with vindesine-ifosfamide- quantitative single photon emission computed tomography
platinum (VIP) chemotherapy followed by platinum sensi- into radiation therapy treatment planning for lung cancer:
tized radiotherapy in stage IIIB non-small cell lung cancer: impact of attenuation and scatter correction on the single
a phase II trial. Lung Cancer 22:45–53 photon emission computed tomography–weighted mean
Van Dyk J, Keane TJ, Kan S et al (1981) Radiation dose and functional lung segmentation. Int J Radiat Oncol
pneumonitis following large single dose irradiation: a re- Bio Phys 78:587–594
evaluation based on absolute dose to lung. Int J Radiat Yom SS, Liao Z, Liu HH et al (2007) Initial evaluation of
Oncol Biol Phys 7:461–467 treatment-related pneumonitis in advanced-stage non–
van Loon J, De Ruysscher D, Wanders R et al (2010) Selective small-cell lung cancer patients treated with concurrent
nodal irradiation on basis of (18)FDG-PET scans in limited- chemotherapy and intensity-modulated radiotherapy. Int J
disease small-cell lung cancer: a prospective study. Int J Radiat Oncol Bio Phys 68:94–102
Radiat Oncol Biol Phys 77:329–336 Yorke ED, Jackson A, Rosenzweig KE, Et al. (2002a) Dose-
van Rijswijk RE, Verbeek J, Haanen C et al (1987) Major volume factors contributing to the incidence of radiation
complications and causes of death in patients treated for pneumonitis in non-small-cell lung cancer patients treated
Hodgkin’s disease. J Clin Oncol 5:1624–1633 with three-dimensional conformal radiation therapy. Int J
Vogt HG, Kolotas C, Martin T et al (1996) Simultaneous Radiat Oncol Biol Phys 54:329–339
radiochemotherapy with paclitaxel in non-small cell lung Yorke ED, Jackson A, Rosenzweig KE (2002b) Dose-volume
cancer: a clinical phase I study. Semin Oncol 23:26–30 factors contributing to the incidence of radiation pneumo-
Wasserman T (1999) Radioprotective effects of amifostine. nitis in non-small-cell lung cancer patients treated with
Semin Oncol 26:89–94 three-dimensional conformal radiation therapy. Int J Radiat
Watchie J, Coleman CN, Raffin TA et al (1987) Minimal long- Oncol Biol Phys 54:329–339
term cardiopulmonary dysfunction following treatment for Yorke ED, Wang L, Rosenzweig KE et al (2002c) Evaluation
Hodgkin’s disease. Int J Radiat Oncol Biol Phys of deep inspiration breath-hold lung treatment plans with
13:517–524 Monte Carlo dose calculation. Int J Radiat Oncol Biol Phys
Werner-Wasik M, Scott C, Movsas B et al (2003) Amifostine as 53:1058–1070
mucosal protectant in patients with locally advanced non- Yu X, Prosnitz RR, Zhou S et al (2003) Symptomatic cardiac
small cell lung cancer (NSCLC) receiving intensive events following radiation therapy for left-sided breast
chemotherapy and thoracic radiotherapy (RT): results of cancer: possible association with radiation therapy-induced
the Radiation Therapy Oncology Group (RTOG) 98-01 changes in regional perfusion. Clin Breast Cancer
study. Int J Radiat Oncol Biol Phys 57:S216 4:193–197
Whelan TJ, Julian J, Wright J et al (2000) Does locoregional Zhao L, Wang L, Ji W et al (2009) Elevation of plasma TGF-
radiation therapy improve survival in breast cancer? A beta1 during radiation therapy predicts radiation-induced
meta-analysis. J Clin Oncol 18:1220–1229 lung toxicity in patients with non-small-cell lung cancer: a
Willner J, Jost A, Baier K et al (2003) A little to a lot or a lot to combined analysis from Beijing and Michigan. Int J Radiat
a little? An analysis of pneumonitis risk from dose-volume Oncol Biol Phys 74:1385–1390
Spinal Cord Toxicity
Timothy E. Schultheiss

Contents Abstract
Radiation myelopathy is a feared and generally
1 Introduction.............................................................. 627 avoidable complication of thoracic irradiation.
2 Signs and Symptoms ............................................... 628 A better understanding of the radiation response
of the spinal cord and advances in radiation
3 Diagnosis ................................................................... 628
therapy delivery techniques mean that this com-
4 Pathology .................................................................. 629 plication should be preventable in nearly all
5 Radiation Protection of the Spinal Cord.............. 630 treatment situations. A better understanding of
6 Dose Limits ............................................................... 630
the pathogenesis of the injury has lead to the
6.1 Differential Sensitivity—Cervical protection of the spinal cord from radiation in
Versus Thoracic......................................................... 630 experimental studies and to the possible treatment
6.2 Fractionation .............................................................. 631 of radiation myelopathy.
6.3 Experimental Studies................................................. 631
6.4 Dose Gradients .......................................................... 632
7 Retreatment .............................................................. 632
8 Chemotherapy .......................................................... 632 1 Introduction
9 Other Factors Affecting Response......................... 633
Thoracic radiation myelopathy does not appear to be
10 Treatment of Radiation Myelopathy..................... 633
pathologically different from cervical radiation mye-
References.......................................................................... 633 lopathy. Fortunately, the morbidity of thoracic radi-
ation myelopathy is less than that of cervical radiation
myelopathy simply because the injury occurs at a
lower level of the spinal cord. However, radiation
myelopathy is still the most feared radiation compli-
cation of lung cancer treatment.
Radiation myelopathy may have been studied more
than any other normal tissue injury with the possible
exception of skin. It has been studied in mice, rats,
cats, guinea pigs, rabbits, pigs, dogs, and monkeys.
These experimental studies have focused primarily on
the pathogenesis, dose response, and fractionation
effects. In the early years of radiation oncology new
T. E. Schultheiss (&) technologies (e.g., introduction of Co-60 and linear
City of Hope National Medical Center,
1500 Duarte Road, Duarte, CA 91010, USA accelerators) and new treatment techniques (e.g.,
e-mail: schultheiss@coh.org hypofractionation, split course treatment, multiple

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 627
DOI: 10.1007/174_2011_274, Ó Springer-Verlag Berlin Heidelberg 2011
628 T. E. Schultheiss

fractions per day) sometimes resulted in an unex- The prognosis with radiation myelopathy is
pectedly high incidence of radiation myelopathy determined by the level of the lesion (Schultheiss
because of unexpected changes in the dose distribu- et al. 1986) and the degree to which the cord is
tion or in the biological dose. Although the field and transected (Holdorff 1980). As with any myelopathy,
our understanding of radiation myelopathy have total cord transection is a poor prognostic sign, and
advanced to the point where a new technique rarely more inferior lesions have a better prognosis.
has an unanticipated risk of radiation myelopathy, the
complication continues to occur. It is now almost
completely confined to mistakes or to the most idio- 3 Diagnosis
syncratic cases, which are by definition difficult to
predict. In the following section, we will try to In an adult, the latency period for radiation myelop-
address the known parameters that influence the athy is rarely less than 6 months from the completion
incidence of thoracic radiation myelopathy as well as of radiation, with no apparent difference between the
the putative factors that could be considered when one cervical and thoracic myelopathies (Chouchair 1991;
may be required to push the spinal cord dose in favor Feldmann and Posner 1986; Schultheiss et al. 1984a).
of tumor control. If a patient’s symptoms date from less than 6 months
post radiation, then one should seek alternative
diagnoses (including unknown causes), determine
2 Signs and Symptoms whether there were excessive doses or mistakes in
dosimetry, or investigate factors that could have
The initial symptoms of radiation myelopathy are combined with the radiation to alter the normal spinal
subtle and nonspecific. Lhermitte sign may precede cord tolerance. Paraneoplastic syndromes sometimes
the development of permanent radiation myelopathy involve the CNS and may have similarities with
by several months. This sign is more frequently seen radiation myelopathy. Furthermore, these syndromes
in a large field or cervical irradiation, but can also are more common in patients with lymphomas and
occur in thoracic irradiation. However, its occurrence lung cancer than most other tumors (Chouchair 1991;
is probably independent of the development of radi- Feldmann and Posner 1986; Schultheiss et al. 1984a).
ation myelopathy since it occurs with radiation A definitive diagnosis of radiation myelopathy
doses well below those that would cause radiation may be difficult to make because of the lack of
myelopathy. It does not predict permanent injury. pathognomonic lesions or findings. Further work up
Symptoms associated with permanent radiation will generally include CT scans, MRI scans, and
myelopathy progress at various rates. The first signs plane films all of which must be negative for tumor or
generally include unilateral paresthesias, numbness, other etiology. Myelograms are rarely performed
clumsiness, lower extremity weakness, and decrease now, but information from cerebrospinal fluid can be
in proprioception. By the time the patient complains useful, especially when neoplastic cells are found in
of symptoms, the symptoms will have intensified in the fluid. Unless an overdosage or predisposing con-
severity and could also include signs and symptoms dition is clearly indicated, there is no logical reason to
such as changes in gait, weakness, hemiparesis, eliminate other causes from the diagnosis of a former
Brown–Sequard syndrome, spasticity, pain, hyperre- radiation patient with myelopathic symptoms. Any
flexia. Babinski signs are common. The signs usually disease affecting the CNS can potentiate a radiation
start in the lower extremities and progress rostrally. injury. Chemotherapy, surgery, and congenital con-
The degree of morbidity can stabilize at almost any ditions may also reduce the spinal cord’s radiation
level, and the ultimate severity has not been shown to tolerance (Abadir 1980; Zulch and Oeser 1974).
be related to the radiation dose. These symptoms are When performed, myelograms are rarely positive
generally characteristic of most demyelinating dis- in radiation myelopathy, although complete blocks
eases except that they are irreversible and do not wax are sometimes seen. Swelling is often noted. Total
and wane. It is not unusual that a traumatic event, protein, myelin basic protein, and lymphocytes in
such as a fall, precedes or initiates neurological the CNS may be elevated (Kitamura et al. 1979;
symptoms. Lechevalier et al. 1973; Marty and Minckler 1973;
Spinal Cord Toxicity 629

Worthington 1979). This could be true of other characterized by collapse of the normal architecture
demyelinating conditions as well. Measurements of and vacuolation (status spongiosis), spheroids, and
nerve conduction velocities show slowed spinal con- glial scars. Mineral deposits may be seen. These
duction or complete blocks (Dorfman et al. 1982; lesions can occur without any clearly associated
Snooks and Swash 1985). changes to the vasculature, with the exception that
Wang et al. (1992) reported ten cases of radiation vascular lesions occur in close association with
myelopathy imaged with magnetic resonance. hemorrhagic necrosis.
Of eight cases examined 2–8 months after onset of Vasculopathies after spinal cord radiation include
symptoms, they noted spinal cord swelling in six the more subtle changes of increased vascularity,
cases and low intensity on T1 weighted images and telangiectasia, and thickening or degeneration of the
high signal intensity on T2 weighted images in eight hyaline. Edema and fibrin exudation are commonly
cases. Diffuse atrophy was found in two cases seen. Perivascular fibrosis, fibrinoid necrosis, throm-
examined 36 and 53 months after onset of symptoms. bosis, and hemorrhage are more severe manifestations
Enhancement with Gd-DTPA contrast is also seen of vascular injury. These changes are found in other
(Alfonso et al. 1997; Koehler et al. 1996; Wang et al. conditions as well.
1992). The inflammatory response is quite variable and
obviously depends on the age of the lesions. Microglia
and astrocytes show the most dramatic changes.
4 Pathology Occasionally there are regions with the normal white
matter completely replaced with microglial macro-
Because the spinal cord has relatively few mechanisms phages and astrocytes. A brisk astrocytic response
it can deploy in response to injury, there are no patho- is a common feature of the irradiated spinal cord and
gnomonic features of radiation injury to the spinal cord. has led to much speculation regarding the role of
Demyelination, white matter necrosis, and malacia are astrocytes in the repair process and as a mediator of
characteristic of radiation injury to the spinal cord. injury.
Histopathologic studies of radiation myelopathy in The pathogenesis of white matter necrosis has been
humans are obviously limited to the autopsy material controversial. Oligodendrocytes and their precursors
(Schultheiss et al. 1988). This results in descriptions of have been viewed as the target cells whose radiation
lesions that are relatively more severe or advanced. death leads ultimately to demyelination and malacia.
Experimental studies provide a more comprehensive Extensive studies have been performed on the iden-
view of these lesions, both in terms of severity and age tification and radiation response of glial progenitor
of the lesion (Black and Kagan 1980; Ruifrok et al. cells (Hornsey et al. 1981; Hubbard and Hopewell
1994; van der Kogel 1974, 1979). 1979; Myers et al. 1986; Otsuka et al. 2006; Philippo
Traditionally, it is common to divide the radiation et al. 2000, 2005; Ruifrok et al. 1994; van der Maazen
injury to the spinal cord into two categories, the white et al. 1990, 1991, 1992, 1993). On the other hand,
matter response and the vascular response. The white endothelial cells have long been suggested as the
matter response includes demyelination of isolated alternative to glial cells as the primary target cells for
nerve fibers and progressing to groups of fibers. radiation injury.
Demyelination can progress to active malacia where Nordal et al. (2004) studied the up-regulation of
the breakdown of the neuropil is ongoing. Active vascular endothelial growth factor (VEGF) in irradi-
malacia will have areas with increased numbers of ated rat and mouse spinal cords. They found rapidly
astrocytes and microglia. These cells perform repair increasing cellular expression of VEGF starting
and phagocytosis, but they may also play a role in the 4 weeks before white matter necrosis and a steep
pathogenesis of the lesion through an increase in the dose–response curve for the expression of VEGF.
production or the release of cytokines such as IL-1 The VEGF expressing cells were identified as astro-
and TNFa (Nordal and Wong 2005; Schultheiss and cytes. Up-regulation of VEGF is associated with
Stephens 1992). Blood may fill a necrotic area increased vascular permeability, edema, and hypoxia
(hemorrhagic necrosis). Inactive malacia may be in the CNS.
630 T. E. Schultheiss

Using boron neutron capture therapy (BNCT) as a irradiated without the drug. After 65 weeks, in
probe for studying CNS radiation injury, Coderre Gammaphos rats the endothelial cell number was
et al. 2006 have shown that the incidence of radiation significantly higher and vasculature abnormalities and
myelopathy does not track the survival of glial pro- necrosis were dramatically lower. This study provides
genitor cells (Morris et al. 1994, 1996, 1997, 1998). increased evidence that injury to the vascular endo-
In these studies, surviving fractions of O2A glial thelium and not glial cells injury leads to white matter
progenitor cells were determined at various times necrosis in the CNS.
after irradiation under conditions where the vascular Following this study, Nieder et al. (2005) treated
endothelium could be preferentially irradiated or the rats with a combination of WR-2721 and insulin-like
entire white matter uniformly irradiated. Very differ- growth factor (IGF) at the time of re-irradiation with
ent surviving fractions of glial progenitor cells were 17–21 Gy. This was given 21 weeks after an initial
seen under irradiation techniques that yielded dose of 16 Gy. The rats receiving IGF showed a
approximately equal incidences of myelopathy and significantly lower myelopathy incidence.
conversely similar surviving fractions were seen
under conditions yielding very different myelopathy
rates. Moreover, the endothelial doses tracked well 6 Dose Limits
with the myelopathy rates.
Thus is it currently believed that the primary target The published ‘‘safe’’ limits of spinal cord dose have
cell giving rise to radiation myelopathy is located in varied over the years. Doses as high as 60 Gy in
the vasculature, presumably the vascular endothelium conventional fraction sizes have been suggested
(Hopewell and van der Kogel 1999). White matter (Baekmark 1975; Kim and Fayos 1981; Verity 1968).
necrosis resulting from oligodendroglial cell death Certainly, 45 Gy is currently the most widely accep-
(with or without the death of its progenitor cell) is no ted dose limit on the spinal cord. However, in practice
longer considered to be a viable hypothesis. Blood– one sees that 45 Gy is often treated as an absolute
brain barrier breakdown, edema, hypoxia, astrocytic dose limit, with lower doses being commonly used.
responses, and alterations of cytokine expression are Clearly, one should not irradiate the spinal cord to
all potentially involved in the pathogenesis (Blake- doses higher than are necessary to achieve the thera-
more and Palmer 1982; Hornsey et al. 1990; Myers peutic goal. However, the converse is also true.
et al. 1986; Nordal and Wong 2005; Schultheiss and Tumor control should not be sacrificed solely to
Stephens 1992). The pathways to injury are not respect a spinal cord tolerance that is unrealistically
completely understood, but strategies for treatment of low.
this injury continue to be developed.

6.1 Differential Sensitivity—Cervical


5 Radiation Protection of the Spinal Versus Thoracic
Cord
Kramer (1968) was the first to publish different dose
Hornsey et al. (1990) found that certain vasoactive limits for the cervical and thoracic spinal cord. They
drugs delayed the expression of ataxia and reduced its were 50 Gy in 5 weeks and 45 Gy in 4 1/2 weeks,
incidence. Their conclusion was that white matter respectively. At this time, it was common to treat with
necrosis was secondary to vessel leakage, edema, one field per day with two opposing fields. Generally
infarction, and transient ischemia and the adminis- the cervical cord was included in lateral head and
tration of these drugs prevented some of the vascular neck fields, and was therefore a midline structure and
injury thereby reducing the consequential white received the same dose each day from the left and
matter injury. right lateral fields. However, the thoracic cord is
In a study by Lyubimova and Hopewell (2004), generally only about 5 cm from the posterior skin and
rats were given Gammaphos, a radioprotector also would have received a high dose on days when the
known as WR-2721 or Amifostine, prior to under- posterior field was treated and a low dose on the days
going brain irradiation. These were compared to rats of anterior field treatment. (Co-60 would have been
Spinal Cord Toxicity 631

the most common teletherapy machine in this time thoracic cord’s tolerance is greater than the cervical
period.) Because the spinal cord is more sensitive to cord. However, this primarily pertains to the high
fraction size than most tissues, the effect of the dose experience. In the lower dose region, the inci-
alternating high and low daily doses was to increase dence is so low that the responses are statistically
the myelopathy risk above that which would have indistinguishable at this time. Generally a lower tol-
been seen if the same total dose had been delivered in erance is respected for pediatric patients (Knowles
constant daily fractions. The result was that the tho- 1983; Schultheiss et al. 1984b), especially when
racic cord was erroneously believed to have a lower radiation is combined with chemotherapy.
tolerance than the cervical cord. The importance of The spinal cord was unexpectedly sensitive to
the individual fraction size was first noted by Marks dose schedules involving multiple fractions per day
et al. in 1973 (Marks et al. 1973). (Dische and Saunders 1989; Wong et al. 1991). This
Forty-five Gy at 1.8–2.0 Gy per day is the most observation resulted in the addition of incomplete
commonly held dose limit. No well documented repair to the LQ model to address the time depen-
myelopathies have been published at this dose level in dence of interfraction repair of radiation damage
the absence of extenuating circumstances. These cir- (Thames 1989; Thames et al. 1988). Marcus and
cumstances are factors in the medical history that Million (1990) reported no cervical myelopathies in
could predispose the patient to radiation injury, one a BID regimen of 1.2 or 1.0 Gy per fraction to total
field per day treatment, neurotoxic chemotherapy, or cord doses of 40–45 Gy in 107 patients or 45–50 Gy
dosimetry errors. Because 45 Gy is on a relatively flat in 90 patients using an interfraction interval of 4–6 h.
portion of the dose–response curve and because the It appears that in BID treatments, a fraction size of
great majority of cases at this dose will have other 1.2 Gy to a dose of 45–50 Gy is safe. The interval
significant contributory factors, it is highly likely that between fractions should be at least 6 h. Because the
cases of myelopathy occurring at 45 Gy would half time of repair and even the validity of first order
also occur at 40 Gy. Thus a miniscule reduction in repair kinetics are uncertain, it would be risky to use
the myelopathy is achieved by routinely using a the LQ model with incomplete repair to extrapolate
lower dose. from a single daily fraction regimen to determine
an equivalent multiple-fractions-per-day regimen.
This risk applies to the clinical use of such a calcu-
6.2 Fractionation lated dose. Dose escalation with hyperfractionation
should be undertaken with caution.
Fractionation schedules and incidences of myelopa-
thy have been collected and published elsewhere
(Schultheiss et al. 1995). Generally, the dose sched- 6.3 Experimental Studies
ules in which more than one occurrence of radia-
tion myelopathy has been seen have employed It is well known that the total doses used in animal
doses per fraction larger than 2 Gy. This is espe- experiments cannot be directly applied to humans. It
cially true for schedules involving thoracic radiation is not known whether this is also true of fraction size,
myelopathy. interfraction interval, relative volume, number of
The dose response of the cervical cord has been fractions, and overall time. This could cast doubts on
shown to fit a logistic function (Schultheiss 2008). the validity of some experimental work that cannot be
The value of a/b of the LQ model was 0.87 Gy. This validated at least in part in humans.
is in reasonable agreement with the available animal Much of the early isoeffect studies in rats seemed
data. The slope of the dose-response function was also to have been in response to clinical observations that
not significantly different from that reported for non- the human spinal cord was more sensitive to increases
human primates (Schultheiss et al. 1990). However, in dose per fraction than was anticipated (Ang et al.
no satisfactory fit was obtained for the thoracic cord. 1983; Hornsey and White 1980; Masuda et al. 1977;
Nearly all of the dose-incidence points for thoracic van der Kogel 1977). This clinical observation was
myelopathy lie to the right of the cervical cord’s partially a result of the experimentation with split
dose-response function. This would indicate that the course radiation schedules, large doses per fraction
632 T. E. Schultheiss

used in hyperbaric oxygen treatments, and hypofrac- volumes of spinal cord, inhomogeneously irradiated.
tionation with insufficient spinal cord shielding. The latency for these cases is consistent with con-
The initial experimental work on radiation mye- ventional cases. Because of the few cases, analysis
lopathy concentrated on fractionation effects. In most of incidence is not practicable. One aspect of these
studies involving the cervical and upper thoracic cases is that they may progress to complete paralysis
levels, the a/b ratio was about 2 Gy (van der Kogel with less certainty than in cases found following
1991). This is not significantly different from the conventional treatments. However, any conclusions
finding in the human cervical cord of 0.87 Gy are preliminary.
(Schultheiss 2008). Careful work by Wong et al.
(1992) found an a/b ratio of 3.4 Gy for rat cord
treated in up to ten fractions, whereas with more than
7 Retreatment
ten fractions they found a/b =0.5 Gy.
Investigators at M. D. Anderson Cancer Center
It is clear that the spinal cord has a significant capacity
performed extensive fractionated studies in rhesus
for recovery from occult damage (Ang et al. 1993,
monkeys using 2.2 Gy fractions (Ang et al. 1993,
2001). van der Kogel (1991) has shown that retreatment
2001; Schultheiss et al. 1990, 1992, 1994; Stephens
tolerance doses increase approximately linearly with
et al. 1983). The treatments were primarily directed at
time after the initial dose, for at least 200 days fol-
the cervical cord. They found that the ED50 ± 1 SE
lowing treatment. Moreover his data show that the
(median tolerance ± standard error) was 76.1 ±
occult damage remaining 200 days after retreatment
1.9 Gy, and the volume effect was consistent with the
increases with the magnitude of the initial dose. The
probability model (Schultheiss et al. 1983). In
greater the initial dose, the less recovery there is. How
humans, no field size effect has been observed with
these data may be used to inform clinicians regarding
respect to incidence, latency, or severity of symptoms.
retreatment in humans is uncertain. The initial dose
However, this is probably a result of too few data
received by a patient in whom retreatment is being
rather than no actual effect.
considered is likely to be very far from tolerance. From
the animal data, this would imply increased recovery.
In general, higher retreatment doses may be given with
6.4 Dose Gradients
lower initial doses and longer intervals between treat-
ments. From the sparse clinical and primate data, it
An important volume effect for modern treatment
appears that at least 50% recovery of 45 Gy would be
technique is the effect of dose fall off across the spinal
obtained 2 years after treatment.
cord. In proton treatments of chordomas and chon-
drosarcomas at Massachusetts General Hospital, the
spinal cord was allowed to receive 53 Gy (equivalent)
to the center of the cord and 64 Gy maximum to the 8 Chemotherapy
surface (Austin et al. 1993). No myelopathies have
been observed. It is difficult to quantify the effect of Most patients receiving thoracic irradiation will have
dose fall off, but it is clear that the reported spinal chemotherapy as part of their treatment regimen.
cord limits should refer to a dose given uniformly Many cytotoxic and cytostatic agents are known to be
across the spinal cord and along a length of several neurotoxic, but the effect of chemotherapy on the
centimeters. The spinal cord can tolerate very small tolerance of the spinal cord is unknown. It is likely
volumes taken to doses that would be unacceptably that there is relatively little effect unless the treat-
high for the entire field length. ments are nearly concurrent or the chemotherapy
There have been few cases of radiation myelop- neurotoxic on its own. Although recommended dose
athy reported following stereotactic radiosurgery limits apply in the absence of chemotherapy treat-
(Gibbs et al. 2009; Ryu et al. 2007; Sahgal et al. ments and chemotherapy may reduce the radiation
2010). These cases are characterized by high doses tolerance, 45 Gy in conventional fraction sizes should
per fraction, very few fractions (1–3), and small still be well tolerated.
Spinal Cord Toxicity 633

Wong et al. (2008) reported success treatment of


9 Other Factors Affecting Response neurological symptoms of a woman with temporal
lobe necrosis following treatment for nasopharyngeal
Treatment in hyperbaric oxygen (HBO) has been cancer extending into the sphenoid sinus. Liu et al.
observed to reduce the tolerance of the thoracic spinal (2009) reported four cases of children treated for
cord to radiation (Coy and Dolman, 1971). In HBO pontine glioma who were suspected to have radiation
treatments for head and neck cancers, van den Brenk necrosis. Three showed improvement clinically and in
et al. (1968) did not find an increase in radiation imaging studies; the fourth was determined to have
myelopathy. In a somewhat related finding, Dische progressive disease rather than necrosis. Two of the
et al. (1986) demonstrated a significant positive effect three cases subsequently had clinical progression, but
of pretreatment hemoglobin level on the incidence whether this was tumor related or due to the necrosis
of radiation myelopathy. Although hypertension is was unclear.
known to reduce radiation tolerance in some organs, Finally, Levin et al. (2010) reported a randomized,
this cannot be confirmed in the spinal cord. There are double-blind placebo-controlled trial of bevacizumab
some experimental data that suggest that this is the for patients with radiation necrosis. All seven patients
case (Asscher and Anson 1962; Hopewell and Wright treated with bevacizumab showed MRI responses and
1970). improvement in neurological symptoms. Five of the
Disease processes and congenital or acquired seven placebo patients showed worsening of symp-
spinal abnormalities, may also effect the radiation toms and two showed only MRI progression. Five of
tolerance of the spinal cord to an unknown degree. the placebo patients crossed over to receive bev-
These effects are anecdotal and their discovery is acizumab after progression. All of these patients
made only after a myelopathy has occurred. If a improved with the treatment.
patient presents severe spinal abnormalities, one There has been one reported case of spinal cord
should attempt to spare the cord as much as possible. infarction after bevacizumab for non-small cell lung
cancer (Masselos et al. 2009). This patient did not
receive any radiation. However, her medical history
10 Treatment of Radiation included epilepsy and hypertension, and she was
Myelopathy being treated with an extensive array of drugs at the
time of presentation. The authors concluded that
Treatment of radiation myelopathy with hyperbaric bevacizumab was not necessarily the sole causative
oxygen or steroids has generally not been successful. agent.
Bevacizumab is a humanized murine monoclonal It is unclear whether radiation myelopathy can be
antibody against VEGF. It is used in the treatment of successfully treated with bevacizumab and such
malignant glioma in combination with radiation ther- treatment does have side effects. However, no con-
apy. Reasoning that upregulation of VEGF may con- sistently successful treatment currently exists. If one
tribute to radiation necrosis in the CNS through were to treat radiation myelopathy, early treatment
breakdown of the blood brain barrier and edema, would be important since lesions can progress to
Gonzalez et al. (2007) suggested that bevacizumab may complete cord transection. As the pathophysiology of
have a beneficial effect against radiation brain necrosis . radiation myelopathy becomes increasingly eluci-
Upon retrospective review of patients treated with dated, we may reasonably expect progress in the
bevacizumab and radiation, they identified 8 patients treatment and prevention of radiation myelopathy.
with radiation brain necrosis diagnosed by MRI and
biopsy confirmed. All 8 patients showed a reduction in
the volume of the imaging abnormalities after bev- References
acizumab treatment for their malignancy. To reiterate,
these cases were found on retrospective review. Abadir R (1980) Radiation myelitis: can diagnosis be unequiv-
ocal with histological evidence? Int J Radiat Oncol Biol
Following this report, two additional studies were Phys 6:649–650
published with patients who had known cerebral Alfonso ER, De Gregorio MA, Mateo P, Esco R, Bascon N,
radiation necrosis and were treated with bevacizumab. Morales F, Bellosta R, Lopez P, Gimeno M, Roca M,
634 T. E. Schultheiss

Villavieja JL (1997) Radiation myelopathy in over-irradi- induced damage to the central nervous system. Front Radiat
ated patients: MR imaging findings. Eur Radiol 7:400–404 Ther Oncol 33:265–275
Ang KK, van der Kogel AJ, van der Schueren E (1983) The Hopewell JW, Wright EA (1970) The nature of latent cerebral
effect of small radiation doses on the rat spinal cord: the irradiation damage and its modification by hypertension. Br
concept of partial tolerance. Int J Radiat Oncol Biol Phys J Radiol 43:161–167
9:1487–1491 Hornsey S, White A (1980) Isoeffect curve for radiation
Ang KK, Price RE, Stephens LC, Jiang GL, Feng Y, myelopathy. Br J Radiol 53:168–169
Schultheiss TE, Peters LJ (1993) The tolerance of primate Hornsey S, Myers R, Coultas PG, Rogers MA, White A (1981)
spinal cord to re-irradiation. Int J Radiat Oncol Biol Phys Turnover of proliferative cells in the spinal cord after X-
25:459–464 irradiation and its relation to time-dependent repair of
Ang KK, Jiang GL, Feng Y, Stephens LC, Tucker SL, Price RE radiation damage. Br J Radiol 54:1081–1085
(2001) Extent and kinetics of recovery of occult spinal cord Hornsey S, Myers S, Jenkinson T (1990) The reduction of
injury. Int J Radiat Oncol Biol Phys 50:1013–1020 radiation damage to the spinal cord by post-irradiation
Asscher AW, Anson SG (1962) Arterial hypertension and administration of vasoactive drugs. Int J Radiat Oncol Biol
irradiation damage to the nervous system. Lancet II: Phys 18:1437–1442
1343–1346 Hubbard BM, Hopewell JW (1979) Changes in the neuroglial
Austin JP, Urie MM, Cardenosa G, Munzenrider JE (1993) cell populations of the rat spinal cord after local X-irradi-
Probable causes of recurrence in patients with chordoma ation. Br J Radiol 52:816–821
and chondrosarcoma of the base of skull and cervical spine. Kim YH, Fayos JV (1981) Radiation tolerance of the cervical
Int J Radiat Oncol Biol Phys 25:439–444 spinal cord. Radiology 139:473–478
Baekmark UB (1975) Neurologic complications after irradia- Kitamura HK, Kameda Y, Yoshimura Y, Magaoka S, Takai S,
tion of the cervical spinal cord for malignant tumour of the Nagatsuka A (1979) Delayed radiation myelopathy. Yoko-
head and neck. Acta Radiol Ther Phys Biol 14:33–41 hama Medical Bulletin 30:61–69
Black MJ, Kagan AR (1980) Transverse myelitis. Laryngo- Knowles JF (1983) The radiosensitivity of the guinea pig spinal
scope 90:847–852 cord to X-rays: the effect of retreatment at 1 year and the effect
Blakemore WF, Palmer AC (1982) Delayed Infraction of of age at the time of irradiation. Int J Radiat Biol 44:433–442
Spinal Cord White Matter Following X-irradiation. J Pathol Koehler PJ, Verbiest H, Jager J, Vecht CJ (1996) Delayed
137:273–280 radiation myelopathy: serial MR-imaging and pathology.
Chouchair AK (1991) Myelopathies in the cancer patient: Clin Neurol Neurosurg 98:197–201
incidence, presentation, diagnosis and management. Oncology Kramer S (1968) The hazards of therapeutic irradiation of the
5:25–37 central nervous system. Clin Neurosurg 15:301–318
Coderre JA, Morris GM, Micca PL, Hopewell JW, Verhagen I, Lechevalier B, Humeau F, Houteville JP (1973) Myelopathies
Kleiboer BJ, van der Kogel AJ (2006) Late effects of radiotherapiques ‘hypertenphiantes’. A propos de cinq
radiation on the central nervous system: role of vascular observations dont une anatome clinique. Revue Neurolog-
endothelial damage and glial stem cell survival. Radiat Res ique 129:119–132
166:495–503 Levin VA, Luc B, Ping H, Ashok JK, Jeffrey SW, Bekele BN,
Coy P, Dolman CL (1971) Radiation myelopathy in relation to Sujit P, Monica L, Mark RG, Edward FJ (2010) Randomized
oxygen level. Br J Radiol 44:705–707 Double-Blind Placebo-Controlled Trial of Bevacizumab
Dische S, Saunders MI (1989) Continuous, hyperfractionated, Therapy for Radiation Necrosis of the Central Nervous
accelerated radiotherapy (CHART): an interim report upon System. Int J Radiat Oncol Biol Phys 79:1487–1495.
late morbidity. Radiother Oncol 16:65–72 Liu AK, Macy ME, Foreman NK (2009) Bevacizumab as
Dische S, Saunders MI, Warburton MF (1986) Hemoglobin, therapy for radiation necrosis in four children with pontine
radiation, morbidity and survival. Int J Radiat Oncol Biol gliomas. Int J Radiat Oncol Biol Phys 75:1148–1154
Phys 12:1335–1337 Lyubimova N, Hopewell JW (2004) Experimental evidence to
Dorfman LS, Donaldson SS, Gupta PR, Bosley TM (1982) support the hypothesis that damage to vascular endothelium
Electrophysiologic evidence of subclinical injury to the plays the primary role in the development of late radiation-
posterior columns of the human spinal cord after therapeutic induced CNS injury. Br J Radiol 77:488–492
radiation. Cancer 50:2815–2819 Marcus RG, Million RR (1990) The incidence of myelitis after
Feldmann E, Posner JB (1986) Episodic neurologic dysfunction in irradiation of the cervical spinal cord. Radiology 93:3–8
patients with Hodgkin’s disease. Arch Neurol 43:1227–1233 Marks RD Jr, Agarwal SK, Constable WC (1973) Increased
Gibbs IC, Patil C, Gerszten PC, Adler JR Jr, Burton SA (2009) rate of complications as a result of treating only one
Delayed radiation-induced myelopathy after spinal radio- prescribed field daily. Radiology 107:615–619
surgery. Neurosurg 64:A67–A72 Marty R, Minckler DS (1973) Radiation myelitis simulating
Gonzalez J, Kumar AJ, Conrad CA, Levin VA (2007) Effect of tumor. Arch Neurol 29:352–354
bevacizumab on radiation necrosis of the brain. Int J Radiat Masselos K, Begbie S, Lees JN (2009) Spinal cord infarction
Oncol Biol Phys 67:323–326 in a patient with metastatic non-small cell lung cancer,
Holdorff B (1980) Dose effect relationships in cervical and receiving chemotherapy combined with bevacizumab. Asia-
thoracic radiation myelopathies. Acta Radiol Oncol 19: Pacific J Clin Oncol 5:151–153
271–277 Masuda K, Reid BO, Withers HR (1977) Dose effect relation-
Hopewell JW, van der Kogel AJ (1999) Pathophysiological ship for epilation and late effects on spinal cord in rates
mechanisms leading to the development of late radiation- exposed to gamma rays. Radiology 122:239–242
Spinal Cord Toxicity 635

Morris GM, Coderre JA, Hopewell JW, Micca PL, Nawrocky Sahgal A, Ma L, Gibbs I, Gerszten PC, Ryu S, Soltys S,
MM, Liu HB, Bywaters A (1994a) Response of the central Weinberg V, Wong S, Chang E, Fowler J, Larson DA
nervous system to boron neutron capture irradiation: (2010) Spinal cord tolerance for stereotactic body radio-
evaluation using rat spinal cord model. Radiother Oncol therapy. Int J Radiat Oncol Biol Phys 77:548–553
32:249–255 Schultheiss TE (2008) The radiation dose-response of the human
Morris GM, Coderre JA, Whitehouse EM, Micca P, Hopewell spinal cord. Int J Radiat Oncol Biol Phys 71:1455–1459
JW (1994b) Boron neutron capture therapy: a guide to the Schultheiss TE, Stephens LC (1992) Permanent Radiation
understanding of the pathogenesis of late radiation damage Myelopathy. Br J Radiol 65:737–753
to the rat spinal cord. Int J Radiat Oncol Biol Phys 28: Schultheiss TE, Orton CG, Peck RA (1983) Models in
1107–1112 radiotherapy: volume effects. Med Phys 10:410–415
Morris GM, Coderre JA, Bywaters A, Whitehouse E, Hopewell Schultheiss TE, Higgins EH, El-Mahdi AM (1984a) The latent
JW (1996) Boron neutron capture irradiation of the rat period in clinical radiation myelopathy. Int J Radiat Oncol
spinal cord: histopathological evidence of a vascular- Biol Phys 10:1109–1115
mediated pathogenesis. Radiat Res 146:313–320 Schultheiss TE, Higgins EM, El-Mahdi AM (1984b) Extrinsic
Morris GM, Coderre JA, Hopewell JW, Rezvani M, Micca PL, versus intrinsic dose dependence of latency in radiation
Fisher CD (1997a) Response of the central nervous system myelopathy. Int J Radiat Oncol Biol Phys 10:2389
to fractionated boron neutron capture irradiation: studies Schultheiss TE, Stephens LC, Peters LJ (1986) Survival in
with borocaptate sodium. Int J Radiat Biol 71:185–192 radiation myelopathy. Int J Radiat Oncol Biol Phys
Morris GM, Coderre JA, Micca PL, Fisher CD, Capala J, 12:1765–1769
Hopewell JW (1997b) Central nervous system tolerance to Schultheiss TE, Stephens LC, Maor MH (1988) Analysis of the
boron neutron capture therapy with p-boronophenylalanine. histopathology of radiation myelopathy. Int J Radiat Oncol
Br J Cancer 76:1623–1629 Biol Phys 14:27–32
Morris GM, Coderre JA, Hopewell JW, Micca PL, Wielopolski Schultheiss TE, Stephens LC, Jiang GL, Ang KK, Peters LJ
L (1998) Boron neutron capture therapy: re-irradiation (1990) Radiation myelopathy in primates treated with
response of the rat spinal cord. Radiother Oncol 48:313–317 conventional fractionation. Int J Radiat Oncol Biol Phys
Myers R, Rogers MA, Hornsey S (1986) A reappraisal of the 19:935–940
roles of glial and vascular elements in the development of Schultheiss TE, Stephens LC, Ang KK, Jardine JH, Peters LJ
white matter necrosis in irradiated rat spinal cord. Br J (1992) Neutron RBE for primate spinal cord treated with
Cancer-Supple 7:221–223 clinical regimens. Radiat Res 129:212–217
Nieder C, Price RE, Rivera B, Andratschke N, Ang KK (2005) Schultheiss TE, Stephens LC, Ang KK, Price RE, Peters LJ
Effects of insulin-like growth factor-1 (IGF-1) and amifos- (1994) Volume effects in rhesus monkey spinal cord. Int J
tine in spinal cord reirradiation. Strahlenther Oncol Radiat Oncol Biol Phys 29:67–72
181:691–695 Schultheiss TE, Kun LE, Ang KK, Stephens LC (1995)
Nordal RA, Wong CS (2005) Molecular targets in radiation- Radiation response of the central nervous system. Int J
induced blood-brain barrier disruption. Int J Radiat Oncol Radiat Oncol Biol Phys 31:1093–1112
Biol Phys 62:279–287 Snooks SJ, Swash M (1985) Motor conduction velocity in the
Nordal RA, Nagy A, Pintilie M, Wong CS (2004) Hypoxia and human spinal cord: slowed conduction in multiple sclerosis
hypoxia-inducible factor-1 target genes in central nervous and radiation myelopathy. J Neurol Neurosurg Psychiatry
system radiation injury: a role for vascular endothelial 48:1135–1139
growth factor. Clin Cancer Res 10:3342–3353 Stephens LC, Hussey DH, Raulston GL, Jardine JH, Gray KN,
Otsuka S, Coderre JA, Micca PL, Morris GM, Hopewell JW, Almond PR (1983) Late effects of 50 MeV neutron and
Rola R, Fike JR (2006) Depletion of neural precursor cobalt-60 irradiation of rhesus monkey cervical spinal cord.
cells after local brain irradiation is due to radiation dose Int J Radiat Oncol Biol Phys 9:859–865
to the parenchyma, not the vasculature. Radiat Res Thames HD (1989) Repair kinetics in tissues: alternative
165:582–591 models. Radiother Oncol 14:321–327
Philippo H, Huiskamp R, Winter AM, Gharbaran B, van der Thames HD, Ang KK, Stewart FA, van der Schueren E (1988)
Kogel AJ (2000) Age dependence of the radiosensitivity of Does incomplete repair explain the apparent failure of
glial progenitors for In vivo fission-neutron and X irradi- the basic LQ model to predict spinal cord and kidney
ation. Radiat Res 154:44–53 responses to low doses per fraction? Int J Radiat Biol
Philippo H, Winter EA, van der Kogel AJ, Huiskamp R (2005) 54:13–19
Recovery capacity of glial progenitors after in vivo fission- van den Brenk HAS, Richter W, Hurley RH (1968) Radiosen-
neutron or X irradiation: age dependence, fractionation and sitivity of the human oxygenated cervical spinal cord based
low-dose-rate irradiations. Radiat Res 163:636–643 on analysis of 357 cases receiving 4 MeV X- rays in
Ruifrok AC, Stephens LC, van der Kogel AJ (1994) Radiation hyperbaric oxygen. Br J Radiol 41:205–214
response of the rat cervical spinal cord after irradiation at van der Kogel AJ (1974) Late effects of spinal cord irradiation
different ages: tolerance, latency and pathology. Int J Radiat with 300 kV X-Rays and 15 MeV neutrons. Br J Radiol
Oncol Biol Phys 29:73–79 45:393–398
Ryu S, Jin JY, Jin R, Rock J, Ajlouni M, Movsas B, Rosenblum van der Kogel AJ (1977) Radiation tolerance of the rat spinal
M, Kim JH (2007) Partial volume tolerance of the spinal cord: time-dose relationships. Radiology 122:505–509
cord and complications of single-dose radiosurgery. Cancer van der Kogel AJ (1979) Late effects of radiation on the
109:628–636 spinal cord. Dose-effect relationships and pathogenesis.
636 T. E. Schultheiss

Unpublished Ph.D. Thesis, University of Amsterdam, determined by an in vitro clonogenic assay after in vitro or in
Amsterdam, Holland vivo fractionated irradiation. Int J Radiat Biol 63:661–666
van der Kogel AJ (1991) Central nervous system radiation Verity GL (1968) Tissue tolerance: central nervous system.
injury in small animal models. In: Gutin PH, Leibel SA, Radiology 91:1221–1225
Sheline GE (eds) Radiation Injury to the Nervous System. Wang PY, Shen WC, Jan JS (1992) Magnetic resonance
Raven Press, New York, pp 91–111 imaging in radiation myelopathy. AJNR 13:1049–1055
van der Maazen RW, Verhagen I, van der Kogel AJ (1990) An Wong CS, Van Dyk J, Simpson WJ (1991) Myelopathy
in vitro clonogenic assay to assess radiation damage in rat following hyperfractionated accelerated radiotherapy for
CNS glial progenitor cells. Int J Radiat Biol 58:835–844 anaplastic thyroid carcinoma. Radiother Oncol 20:3–9
van der Maazen RW, Verhagen I, Kleiboer BJ, van der Kogel Wong CS, Minkin S, Hill RP (1992) Linear quadratic model
AJ (1991) Radiosensitivity of glial progenitor cells of the underestimates sparing effect of small doses per fraction in
perinatal and adult rat optic nerve studied by an in vitro rat spinal cord. Radiother Oncol 23:176–184
clonogenic assay. Radiother Oncol 20:258–264 Wong ET, Huberman M, Lu XQ, Mahadevan A (2008)
van der Maazen RW, Verhagen I, Kleiboer BJ, van der Kogel Bevacizumab reverses cerebral radiation necrosis. J Clin
AJ (1992) Repopulation of O-2A progenitor cells after Oncol 26:5649–5650
irradiation of the adult rat optic nerve analyzed by an in Worthington BS (1979) Diffuse cord enlargement in radiation
vitro clonogenic assay. Radiat Res 132:82–86 myelopathy. Clin Radiol 30:117–119
van der Maazen RW, Kleiboer BJ, Verhagen I, van der Kogel AJ Zulch KJ, Oeser H (1974) Delayed spinal radionecrosis-a
(1993) Repair capacity of adult rat glial progenitor cells juridical error? Neuroradiology 8:173–176
Radiation Therapy-Related Toxicity:
Esophagus
Voichita Bar Ad and Maria Werner-Wasik

Contents Abstract
Radiation-induced esophagitis is a dose-limiting
1 Pathophysiology and Clinical Picture toxicity of lung cancer treatment. The majority of
of Esophagitis ........................................................... 638 patients receiving concurrent chemotherapy and
2 Evaluation of Esophagitis ....................................... 639 thoracic irradiation experience acute esophagitis.
3 Incidence of Esophagitis and Predisposing Acute esophagitis may be disabling and necessitate
Factors....................................................................... 639 hospitalization, placement of a feeding tube in the
stomach, or initiation of parenteral nutrition.
4 Dosimetric Factors Associated
with Esophagitis ....................................................... 641 Moreover, interruption of the course of radiation
therapy may be required in order to permit healing
5 Strategies Used to Prevent or Treat
Esophagitis................................................................ 643 of the esophageal injury. Such treatment breaks
have been demonstrated to decrease survival of
References.......................................................................... 644
patients with unresectable lung cancer. Proper
prevention, diagnosis, and treatment of esophagitis
are therefore essential, as it may have a direct
influence on tumor control and survival.

Abbreviations

LD Lethal dose
NCI CTCAE National Cancer Institute’s Common
Terminology Criteria for Adverse
Events
RTOG Radiation Therapy Oncology Group
CHART Continuous hyperfractionated accel-
erated radiation therapy
EORTC European Organization for Research
and Treatment of Cancer
V. Bar Ad (&)  M. Werner-Wasik 3DCRT Three-dimensional conformal radia-
Department of Radiation Oncology, tion therapy
Kimmel Cancer Center of Jefferson Medical College, DVHs Dose–volume histograms
Thomas Jefferson University Hospital,
QUANTEC Quantitative analysis of normal tissue
111 South 11th Street, Philadelphia,
PA 19107, USA effects in the clinic
e-mail: Voichita.Bar-Ad@Jeffersonhospital.org CT Computed tomography

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 637
DOI: 10.1007/174_2011_204, Ó Springer-Verlag Berlin Heidelberg 2011
638 V. Bar Ad and M. Werner-Wasik

MTD Maximal tolerated dose The first symptoms of acute esophagitis usually
Vx Volume receiving more than x Gy start in the second or third week of thoracic radiation
SBRT Stereotactic body radiation therapy therapy, corresponding to a dose of 18.0–21.0 Gy of
TITE-CRM Time-to-event continual reassessment standard fractionated radiotherapy, and include a
method sensation of difficult swallowing (dysphagia). This
DLTs Dose-limiting toxicities may progress to painful swallowing of food and saliva
TD Tolerance dose (odynophagia) and later to constant pain not neces-
IMRT Intensity modulated radiation therapy sarily related to the swallowing act. In severe cases,
NTCP Normal tissue complication patients may not be able to swallow at all. In patients
probability receiving concurrent chemotherapy and thoracic
CGE Cobalt-gray equivalent radiotherapy, maximal symptoms of acute esophagitis
CCT Concurrent chemotherapy developed within 1, 2, and 3 months from the start of
radiotherapy in 19, 32, and 33% of the patients,
respectively (Werner-Wasik et al. in press).
Patients with esophagitis require steady supportive
1 Pathophysiology and Clinical care, starting with a low-acid and bland diet when the
Picture of Esophagitis first sensation of difficulty swallowing is reported.
Patients should be instructed to avoid coffee, hot bev-
The esophagus is lined with a convoluted squamous erages, spicy foods, citrus fruit and juices, tomato
epithelium, with a basal cell layer, submucosa and a products, alcohol, and tobacco. In addition, a mixture
layer of striated muscle fibers underneath and without of a local anesthetic (2% viscous lidocaine), coating
surrounding serosa. In mice treated with a single fraction substance (benadryl elixir), and saline/baking soda
of radiation therapy to the thorax, evidence of damage to (‘‘Magic Mouthwash’’) is frequently prescribed to be
the esophagus was observed at a dose of 20.0 Gy, taken liberally before meals to facilitate swallowing.
starting 3 days after radiotherapy (Phillips and Ross Once symptoms progress (e.g., pain is more severe and
1974). This included vacuolization of the basal cell only a soft diet is feasible), stronger oral analgesic
layer, absence of mitosis, and submucosal edema. Some agents should be instituted (hydrocodone with acet-
regeneration was evident by 1–2 weeks from radio- aminophen; liquid morphine; prolonged action opiate
therapy, including proliferating basal cells, regenerating preparations, etc.) to control pain and allow adequate
epithelium, and scattered areas of complete esophageal oral nutrition. High-calorie liquid oral nutritional sup-
denudation. At 3 weeks, the regeneration of the esoph- plements are helpful in maintaining satisfactory caloric
ageal lining was complete, and after 4 weeks the intake and minimizing weight loss and anemia. Once
appearance of the irradiated esophagus was normal. For adequate oral intake of fluids is impaired (as deter-
fractionated radiotherapy doses, the LD50/28 (or radio- mined by dietary interview, positional changes in
therapy dose causing death of 50% of the animals over blood pressure and low urinary output), intravenous
28 days) was estimated as 57.45 Gy (in ten fractions). fluids should be instituted promptly in order to break
Radiological findings of esophageal injury were the vicious cycle of dehydration-poor oral intake-more
described in 30 symptomatic patients who received dehydration. A simple initial step is to give fluids
thoracic radiotherapy to 45–60 Gy. The most common intravenously on an outpatient basis for a day or two
finding was esophageal dysmotility, such as failure to while continuing thoracic radiotherapy. When the
complete primary peristaltic waves, non-peristaltic or patient is unable to swallow despite optimal oral
tertiary contractions, or failure of distal esophageal analgesics, hospitalization is indicated for intravenous
sphincter relaxation. Smooth esophageal strictures hydration and intravenous pain control. In extreme
were demonstrated in some patients, and one frank cases, placement of a gastric tube, parenteral nutrition,
ulceration of the irradiated site was observed (Goldstein or urgent operative intervention may be necessary. The
et al. 1975). Abnormal esophageal motility was noted to speed of recovery from acute esophagitis seems related
occur within 4–12 weeks from radiotherapy alone and to the recovery from neutropenia induced by concur-
as early as after 1 week of concurrent chemotherapy rent delivery of chemotherapy. Prolonged neutropenia
and radiotherapy (Lepke and Libshitz 1983). prevents sufficient healing of the esophageal mucosa.
Radiation Therapy-Related Toxicity: Esophagus 639

Table 1 Version 4.0 of National Cancer Institute’s Common Terminology Criteria for Adverse Events
Grade 1 Grade 2 Grade 3 Grade 4 Grade
5
Dysphagia Symptomatic, able to eat Symptomatic and Severely altered eating/ Life-threatening Death
a regular diet altered eating/ swallowing; tube consequences;
swallowing feedings or TPN or urgent intervention
hospitalization indicated indicated
Esophagitis Asymptomatic; clinical Symptomatic; Same as above Life-threatening Death
or diagnostic altered eating/ consequences;
observations only; swallowing; oral urgent operative
intervention not supplements intervention
indicated indicated indicated
TPN total parenteral nutrition

This is a classic indication for a temporary suspension Criteria for Adverse Events (v4.0, CTCAE scale)
of radiotherapy and administration of a granulocyte- as a disorder characterized by inflammation of the
stimulating factor preparation in order to shorten the esophageal wall, and its grading is predominantly
neutropenic period. Short of this, thoracic radiotherapy based on symptoms, altered diet, and need for inter-
should be continued as long as clinical judgment vention (Table 1).
allows, since radiotherapy breaks are strongly associ- Grading systems such as the NCI CTCAE describe
ated with decreased chances of tumor control (Cox toxicity at one point in time, but they do not provide
et al. 1993). information about the length of time over which the
Symptoms of acute esophagitis commonly persist patient experiences the symptoms of esophagitis.
for 1–3 weeks after the completion of radiotherapy. Esophagitis index (Werner-Wasik et al. 2002) is another
Late esophageal damage may subsequently develop at measure of toxicity that is obtained by plotting the
3–8 months from completion of radiotherapy. It most esophagitis grade over time. It may be a more compre-
often manifests as dysphagia to solids, caused by a hensive measure of normal tissue toxicity than maxi-
permanent narrowing of the esophagus (stricture). mum grade alone. Its calculation requires prospective
The presence of stricture requires periodic surgical accumulation of data points of toxicity over time, how-
dilation of the esophagus, usually with excellent ever, and its applicability may therefore be limited to
results (Choi et al. 2005). investigational pursuits. The Radiation Therapy Oncol-
Acute esophagitis may be disabling, resulting in ogy Group (RTOG) 98-01 study (Movsas et al. 2005)
hospitalization, placement of a feeding tube in the implemented other measures of esophagitis, based on
stomach, or parenteral nutrition for a period of time. physician assessment (weekly physician dysphagia log)
Additionally, the course of radiotherapy may need to be as well as daily patient assessment of their difficulty in
halted temporarily in order to allow for healing of the swallowing (patient swallowing diary). These measures
esophageal lining. Treatment breaks in turn have been allow a direct comparison of health care worker- vs.
unequivocally demonstrated to decrease survival of patient-reported outcome endpoints.
patients with unresectable lung cancer (Cox et al. 1993).
Steps toward prevention, prompt diagnosis, and effective
treatment of radiation esophagitis may therefore have a 3 Incidence of Esophagitis
direct impact on tumor control and overall survival. and Predisposing Factors

The reported incidence of severe acute esophagitis


2 Evaluation of Esophagitis (Grade [3) in patients treated with standard thoracic
radiotherapy alone is 1.3% (Werner-Wasik et al. in
Various criteria have been used to grade acute press). Induction chemotherapy increases this risk
esophagitis. ‘‘Esophagitis’’ is defined in Version 4.0 slightly (Byhardt et al. 1998). Only 6% of patients
of National Cancer Institute’s Common Terminology receiving induction chemotherapy followed by
640 V. Bar Ad and M. Werner-Wasik

Table 2 RTOG/EORTC late esophagitis criteria


Grade 0 Grade 1 Grade 2 Grade 3 Grade 4
Esophagus No Mild fibrosis; slight Unable to take solid food Severe fibrosis; able to Necrosis/
symptoms difficulty in normally; swallowing swallow only liquids; may perforation
swallowing solids; no semi-solid food; dilation have pain on swallowing; fistula
pain on swallowing may be indicated dilation required
RTOG/EORTC Radiation Therapy Oncology Group/European Organization for Research and Treatment of Cancer

standard radiotherapy developed severe acute concurrently. Vokes et al. (2002) described an inci-
esophagitis. When chemotherapy is given concur- dence of 52% of severe acute esophagitis (35% Grade
rently with thoracic radiotherapy, however, a strong 3 and 17% Grade 4 esophagitis) with concurrent
radiosensitizing effect is evident. In the RTOG gemcitabine and thoracic radiotherapy. Pulsed low-
experience, 95% of patients receiving concurrent dose paclitaxel (at escalating doses of 15, 20, and
chemotherapy and thoracic radiotherapy experienced 25 mg/m2 infused on Monday, Wednesday, and Fri-
some degree of acute esophagitis. The highest grade day, respectively) delivered concurrently with daily
of esophagitis reported was Grade 1 in 20% of chest irradiation for radiosensitization was associated
patients, Grade 2 in 41%, Grade 3 in 31%, and with 17% risk of Grade 3 esophagitis; no Grade 4 or 5
Grade 4 in 2%. The incidence of severe esophagitis esophagitis was reported in this study (Chen et al.
was higher (70 vs. 22%; p \ 0.0001) in patients 2008). Other study used weekly paclitaxel 60 mg/m2
receiving hyperfractionated radiotherapy than in concurrently with standard fractionated radiotherapy;
patients treated with conventional fractionation the major toxicity was esophagitis, with 20% of the
(Werner-Wasik et al. in press). In other series, patients developing Grade 4 esophagitis (Choy and
altered fractionation has been found to worsen the Safran 1995). These experiences indicate that it is
severity and duration of esophagitis. Ball et al. difficult to predict the severity of esophageal toxicity
(1995) demonstrated that the duration of symptom- a novel chemoradiation regimen will cause. Whether
atic esophagitis was 1.4 months in the conventional the degree of esophagitis is related to scheduling of
radiotherapy arm, 1.6 months in the arm given chemotherapy used (daily vs. weekly vs. every three
conventional radiotherapy with concurrent carbo- weeks) is uncertain.
platin, 3.2 months in the accelerated radiotherapy Other factors associated with a higher risk of
arm, and 2.4 months in the arm where accelerated esophagitis include age [70 years (Langer et al.
radiotherapy was combined with concurrent carbo- 2001), presence of dysphagia before radiotherapy
platin. With the continuous hyperfractionated accel- initiation (Ahn et al. 2005), low pre-treatment body
erated radiation therapy (CHART) regimen, used mass index (Patel et al. 2004), and nodal stage of N2
without chemotherapy for locally advanced non- or worse (Ahn et al. 2005). The extent of nodal
small-cell lung cancer, 19% of patients experienced involvement probably serves as a surrogate for the
severe esophagitis (Saunders et al. 1997). Another volume of esophagus irradiated.
altered fractionation scheme, the concomitant boost Despite high rates of acute esophagitis, late
technique, resulted in a dose-limiting incidence of esophageal damage was infrequent (2% incidence) in
severe esophagitis of 33% of patients when delivered the RTOG experience. Death due to esophagitis was
concurrently with chemotherapy (Dubray et al. practically non-existent (Werner-Wasik et al. in
1995). press). Ahn et al. (2005) showed that the presence of
It had been reported that particular agents, such as acute esophageal injury was the most predictive factor
doxorubicin, cause severe primary or recall esopha- for the development of late esophageal toxicity
gitis at radiotherapy doses as low as 20.0 Gy (Boal (stricture or fistula). The RTOG/EORTC (Radiation
et al. 1979). Gandara et al. (2003) reported a Therapy Oncology Group/European Organization for
20% incidence of severe acute esophagitis with Research and Treatment of Cancer) criteria for late
radiotherapy using the cisplatin/etoposide regimen esophagitis are presented in Table 2.
Radiation Therapy-Related Toxicity: Esophagus 641

4 Dosimetric Factors Associated


with Esophagitis

Preclinical data suggest that doubling the length of


irradiated portion of the esophagus leads to a decrease
of the LD50 (dose causing the death of 50% of irra-
diated animals) (Michalowski and Hornsey 1986).
However, the clinical evidence that esophageal tox-
icity is correlated to esophageal length irradiated
remains controversial. Ball et al. (1995) analyzed the
outcomes of 100 patients divided into three groups
based on length of treatment field (\14.0, 14.0–15.9,
and [16.0 cm) presumed to correlate with the length Fig. 1 Incidence of acute esophagitis according to Vx (volume
of esophagus irradiated. No relationship between receiving more than x Gy). X-axis values estimated according to
treatment field length and the severity of esophagitis range of doses reported. Each curve annotated as follows: Vdose
(investigator, number of patients, percentage with concurrent
was observed. In Choy’s analysis of 120 patients chemotherapy [CCT]. Percentage of patients who received
(Choy et al. 1999), there was no correlation between sequential chemotherapy in in studies by Ahn et al. (2005),
esophagitis grade and length of esophagus in either Belderbos et al. (2005), and Kim et al. (2005) was 44, 38, and
the primary (p = 0.4) or boost (p = 0.1) radiation 15%, respectively. Data for V50 (Ahn et al. 2005) at 15, 45, and
75 Gy represent reported rates of Grade 2 or greater acute
fields. We studied 105 lung cancer patients treated esophagitis plotted in dose bins at \30, 30–60, and [60%,
with concurrent chemoradiotherapy or radiotherapy respectively. Similarly, for V70 (Ahn et al. 2005), V50
alone. Acute esophagitis was scored prospectively in (Rodriguez et al. 2005), and V60 (Kim et al. 2005), each
a uniform fashion, and precise measurements of the symbol represents rates of acute esophagitis at \10% versus
11–30% versus 31–64%, and\30 versus[30%, and\30 versus
esophageal length within each radiation field were [30%, respectively. Dashed horizontal lines reflect dose ranges
available (Werner-Wasik et al. 2000). Again, ascribed to each data point. Upper x-axis range of greatest data
increasing length of esophagus in the radiation field point for V50 (Rodriguez et al. 2005), V50 (Ahn et al. 2005),
did not predict for the severity of acute esophagitis. and V60 (Kim et al. 2005), are indefinite according to data
(light-gray dotted bars). Solid and open symbols represent
Recent advances in three-dimensional conformal reported rates of Grade 2 or greater acute esophagitis and Grade
radiation therapy (3DCRT) allows us to correlate vol- 3 or greater acute esophagitis, respectively. Thicker and thinner
umetric data to organ damage rather than rely on the solid lines represent higher and lower doses of Vx, respectively
older estimates based on organ length (esophagus). The (i.e., thicker line for V70 and thinner line for V20). Reprinted
from Werner-Wasik et al. (2010). Copyright (2010), with
dosimetric study by Maguire et al. (1999) established a permission from Elsevier
relationship between high dose irradiation of the entire
esophageal circumference and esophagitis risk. They
reported a detailed dosimetric analysis of 91 patients in the clinic (QUANTEC) group (Werner-Wasik et al.
treated to a median corrected dose of 78.8 Gy. The 2010). In general, the data are consistent with some
percent of esophageal volume treated to[50.0 Gy and risk of acute esophagitis at intermediate doses of 30–
the maximum percent of esophageal circumference 50 Gy and an increasing effect for greater doses.
treated to[80.0 Gy were significant predictors of late Volumes of esophagus receiving doses higher than
(but interestingly not acute) esophagitis. The concept 50 Gy have been identified as highly statistically
emerging from this data was the importance of sparing significantly correlated with acute esophagitis in
portions of the esophageal circumference to limit several studies. Rates of acute Grade [2 esophagitis
esophageal toxicity. appear to increase to over 30% as V70 exceeds 20%,
The association between esophageal dose–volume V50 exceeds 40%, and V35 exceeds 50% (Fig. 1).
histograms (DVHs) and the risk of radiation-induced However, due to diverse methods of reporting volu-
esophagitis was further evaluated by several authors. metric data (absolute volume or area, relative volume
These data were recently reviewed by the organ or area, and circumferential measures), no consensus
specific quantitative analysis of normal tissue effects for definitive dosimetric recommendations has been
642 V. Bar Ad and M. Werner-Wasik

reached. Another confounding factor is the fact that in response relationship. In the currently ongoing RTOG
some studies the entire esophagus was not delineated, 0617 Phase III comparison of 60 vs. 74 Gy thoracic
making volumetric measurements difficult to inter- radiotherapy with concurrent and consolidation pac-
pret (Werner-Wasik et al. 2010). The entire length litaxel, carboplatin chemotherapy ± cetuximab in
has to be identified (from the crycoid cartilage to patients with stage IIIA/IIIB non-small-cell lung
the gastro-esophageal junction). Moreover, on axial cancer, a mean esophageal dose of\ 34 Gy is strongly
computed-tomography (CT) imaging, the esophageal recommended, but not absolutely required. The
circumference varies significantly. This is a reflection esophageal V60 (volume of the esophagus exceeding
of the swallowing act, and it does not reflect the ana- 60 Gy) should be calculated for each patient.
tomic reality of a relatively uniform circumference Hypofractionated radiotherapy for centrally loca-
(Kahn et al. 2005). Therefore, conventional DVHs ted lesions may expose parts of the esophagus to large
might not correctly reflect the partial volume dose and doses per fraction. The clinical experience using this
may be misleading. Likewise, the esophagus is treatment approach for non-small-cell lung cancer is
slightly mobile; the cephalad, middle, and caudal very limited. The ongoing RTOG 0813 Phase I/II
esophagus can move \ 5, 7, and 9 mm, respectively study is a radiotherapy dose escalation trial using
during normal respiration (Dieleman et al. 2007). This stereotactic lung radiotherapy for early stage, cen-
may introduce additional uncertainties into volumetric trally located, non-small-cell lung cancer in medically
analyses based on planning CT scans. No specific inoperable patients. The primary end point of the trial
margin recommendations can be given at the present is to determine the maximal tolerated dose (MTD) of
time (Werner-Wasik et al. 2010). stereotactic body radiation therapy (SBRT) delivered
Recent studies evaluating escalating radiotherapy in five fractions over 1.5–2 weeks of treatment. The
dose (standard fractionation) or hypofractionated maximum dose aimed to be reached in this trial is
schedules for lung cancer have been published or 60 Gy using 12 Gy per fraction over 1.5–2 weeks.
designed. In each situation, novel restraints had to be Doses will be allocated to the enrolled patients by
placed on the doses to be delivered to the normal organs using the time-to-event continual reassessment
in the chest such as lung, esophagus, and spinal cord. method (TITE-CRM) rather than the conventional
RTOG 93-11 study evaluated the feasibility of Phase I design of 3 patients for each dose level. The
dose escalation for patients with locally advanced first patients are treated with 50 Gy delivered using
non-small-cell lung cancer treated with 3DCRT to the 10 Gy per fraction over 1.5–2 weeks. Dose levels for
gross tumor only, without elective nodal irradiation. subsequent patients enrolled in this study will be
Maximum doses of 77.4–90.3 Gy were prescribed, determined based on the dose limiting toxicities
depending on the percentage of the total lung (DLTs) to the critical organs experienced in previous
receiving more than 20.0 Gy. This trial of thoracic patients. Moreover, a patient may not be assigned to a
radiotherapy alone escalated the dose to the tumor up next higher dose level until there is at least 1 year of
to 90.3 Gy in group 1 (\25% of both lungs receiving cumulative observation at the ‘‘in progress’’ dose
[20 Gy), up to 83.8 Gy in group 2 (25–37% of lungs level. As a result, more patients will be spared DLTs
receiving [20 Gy) and up to 77.4 Gy in group 3 and more patients will be treated at a dose level that
([37% of lung receiving [20 Gy). The maximum ultimately will be selected as the most appropriate
dose allowed to 1/3 of esophageal volume was 65 Gy; one. The maximum point dose to the esophagus
to 2/3 of the volume, 58 Gy; and to the whole accepted is 105% of the target prescription; it is
esophagus, 55 Gy, respectively. The clinical endpoint recommended that less than 5 cc of the esophagus
was esophageal stricture or perforation. No severe (non-adjacent wall) will receive a maximum dose of
acute esophagitis was observed even in the highest less than 27.5 Gy (5.5 Gy per fraction).
radiotherapy dose level. However, the estimated rate The existing models and dose–volume parameters
of late Grade [3 esophageal toxicity at 18 months should be applied only to regimens using conven-
was 8, 0, 4, and 6% for group 1 patients receiving tional fractionated radiotherapy regimens using
70.9, 77.4, 83.8, and 90.3 Gy,respectively and 0 and between 30 and 35 fractions. A prospective assess-
5%, respectively for group 2 patients receiving 70.9 ment of the dose–volume parameters related to
and 77.4 Gy (Bradley et al. 2005), suggesting a dose– esophageal toxicity associated with hypofractionated
Radiation Therapy-Related Toxicity: Esophagus 643

schedules of radiotherapy is required, given the volume and essentially zero deep to it. As a result,
growing interest in this radiation therapy treatment dose to the tumor can be increased, while the fre-
approach. quency and severity of the treatment-related toxicity
Compared to acute esophagitis, late esophageal could theoretically be decreased. This suggests a
toxicity is far less common. The TD5/5 (tolerance significant potential for therapeutic gains (Suit et al.
doses for the esophagus causing clinical stricture or 2003).
perforation in 5% of irradiated patients at 5 years) are Chang et al. (2009) analyzed data on 42 patients
quoted as 60 Gy for the entire esophagus, 58 Gy for diagnosed with stage III non-small-cell lung cancer
two-thirds of the organ and 55 Gy for one-third of the treated with thoracic radiotherapy using proton beam
esophagus (Emami 1996). therapy to a total dose of 74 CGE (Cobalt-Gray
Equivalent) with concurrent carboplatin and paclit-
axel. Esophagitis was reported in only 6.7% of the
5 Strategies Used to Prevent or Treat cases. However, clinical experience with proton
Esophagitis radiation therapy for non-small-cell lung cancer has
been rather limited to date.
Up to now, standard radiotherapy techniques have not Complete exclusion of the esophagus from the
been able to lower the maximum radiotherapy doses to standard radiotherapy field designed to treat a locally
the esophagus significantly. Intensity modulated advanced lung cancer is most often not feasible due to
radiation therapy (IMRT) seems well suited for such a the organ’s central position in the mediastinum.
purpose, with its ability to deliver concave-shaped Therefore, strategies to limit esophagitis using a
radiotherapy dose distributions around organs at risk, radioprotective agent have been explored.
such as esophagus. Grills et al. (2003) compared four A number of clinical trials have investigated the
different radiotherapy techniques for 18 patients with use of amifostine as a radioprotective agent with
stage I–IIIB inoperable non-small-cell lung cancer: thoracic irradiation for lung cancer. Encouraging
IMRT, optimized 3DCRT using multiple beam angles, results reported in Phase II (Komaki et al. 2004) as
limited 3DCRT using 2–3 beams, and traditional well as Phase III randomized trials (Antonadou et al.
radiotherapy using elective nodal irradiation to treat 2001) led to the development of RTOG 98-01, a large
the mediastinum. The techniques were compared by cooperative group Phase III randomized study that
giving each plan a tumor control probability equiva- tested the efficacy of amifostine in preventing
lent to that of the optimized 3DCRT plan delivering esophagitis in non-small-cell lung cancer patients
70 Gy. Using this method, IMRT and 3DCRT offered receiving thoracic radiotherapy with concur-
similar results with regard to the mean lung dose and rent carboplatin, paclitaxel chemotherapy (Movsas
the esophageal normal-tissue complication probability et al. 2005). Patients were randomized to receive
(NTCP) in lymph node negative patients. However, amifostine (500 mg intravenously four times weekly,
IMRT reduced the lung V20 and the mean lung dose preceding the afternoon dose of radiotherapy) versus
by approximately 15% and the lung NTCP by 30% no amifostine. RTOG 98-01 showed that amifostine
when compared with 3DCRT in lymph node positive did not reduce severe esophagitis (30% rate with
cases. The authors concluded that IMRT is beneficial amifostine vs. 34% without), as assessed by the NCI
in selected patients, particularly those with positive CTCAE criteria and weekly physician dysphagia logs.
lymph nodes or those with target volumes in close Evaluation of patient diaries, however, indicated that
proximity to the esophagus. Moreover, in lymph node amifostine conferred a significant reduction in swal-
positive patients, IMRT allowed delivery of radiation lowing dysfunction measured over time (equivalent of
doses 25–30% greater than 3DCRT while maintaining esophagitis index), a decrease in pain after chemora-
an equal probability of pulmonary and esophageal diotherapy, and diminished weight loss in patients
toxicity. receiving chemoradiation (Sarna et al. 2008). Of note,
Proton beam radiotherapy allows a rapidly only 40% of all radiotherapy fractions were ‘‘pro-
increasing dose at the end of the beam range (Bragg tected’’ by amifostine infusion in that study, and only
peak). Therefore, with proper distribution of proton 29% of patients received the medication according
energies the dose can be uniform across the target to the protocol. Further investigation of this agent
644 V. Bar Ad and M. Werner-Wasik

is therefore justified, possibly with subcutaneous Boal DK, Newburger PE, Teele RL (1979) Esophagitis induced
administration in order to increase the compliance and by combined radiation and adriamycin. Am J Radiol 132:
567–570
to allow higher dose intensity of amifostine. Bradley J, Graham MV, Winter K et al (2005) Toxicity and
Another agent utilized to limit esophagitis is oral outcome results of RTOG 9311: a phase I-II dose escalation
sucralfate. Although applied commonly in the clinic, study using three-dimensional conformal radiation therapy
this agent was shown not to have value in decreasing in patients with inoperable non-small-cell lung carcinoma.
Int J Radiat Oncol Biol Phys 61:318–328
acute esophagitis in a double-blind Phase III ran- Byhardt RW, Scott C, Sause WT et al (1998) Response,
domized trial of 97 patients receiving thoracic toxicity, failure patterns, and survival in five RTOG trials of
radiotherapy (McGinnis et al. 1997). sequential and/or concurrent chemotherapy and radiother-
An interesting approach of plasmid/liposome apy for locally advanced non-small cell carcinoma of the
lung. Int J Radiat Oncol Biol Phys 42:469–478
delivery by the human manganese superoxide dis- Chang JY, Komaki R, Bucci MK et al (2009) Failure patterns
mutase transgene has been reported to be successful and toxicity of concurrent proton therapy and chemotherapy
in prevention of radiation esophagitis in mice for stage III non-small cell lung cancer. Int J Radiat Oncol
receiving carboplatin, paclitaxel, and thoracic radio- Biol Phys 75(3):S446
Chen Y, Pandya KJ, Feins R et al (2008) Toxicity profile and
therapy (Stickle et al. 1999). pharmacokinetic study of a phase I low-dose schedule-
Palifermin is a human recombinant keratinocyte dependent radiosensitizing paclitaxel chemoradiation regi-
growth factor shown to significantly reduce severe men for inoperable non-small cell lung cancer. Int J Radiat
oral mucositis in patients with hematological malig- Oncol Biol Phys 71:407–413
Choi GB, Shin JH, Song HY et al (2005) Fluoroscopically
nancies treated with bone marrow transplantation guided balloon dilation for patients with esophageal stric-
(including total body irradiation) (Spielberger et al. ture after radiation treatment. J Vasc Interv Radiol 16(12):
2004). However, the clinical data including patients 1705–1710
with non-hematological tumors treated with radio- Choy H, Safran H (1995) Preliminary analysis of a phase II
study of weekly paclitaxel and concurrent radiation therapy
therapy are limited. for locally advanced non-small cell lung cancer. Semin
In summary, significant progress has been Oncol 4(Suppl 9):55–57
accomplished in our understanding of the basis of Choy H, LaPorte K, Knill-Selby E, Mohr P, Shy Y (1999)
radiation-induced esophageal injury. Addressing this Esophagitis in combined modality therapy for locally
advanced non-small cell lung cancer. Sem Rad Oncol 9:
dose-limiting toxicity is imperative for the intensifi- 90–96
cation of radiotherapy and chemotherapy protocols. Cox JD, Pajak TF, Asbell S et al (1993) Interruptions of high-
Future effort is necessary in order to find effective dose radiation therapy decrease long-term survival of
measures to minimize or eliminate esophagitis. favorable patients with unresectable non-small cell carci-
noma of the lung: analysis of 1244 cases from 3 RTOG
trials. Int J Radiat Oncol Biol Phys 27:493–498
Disclosure No conflict of interest to declare.
Dieleman EMT, Senan S, Vincent A et al (2007) Four-
dimentional computed tomographic analysis of esophageal
mobility during normal respiration. Int J Radiat Oncol Biol
References Phys 67:775–780
Dubray B, Livartowski A, Beuzeboc P, Pouillart P, Cosset JM
(1995) Combined chemoradiation for locally advanced non-
Ahn S, Kahn D, Zhou S et al (2005) Dosimetric and clinical small cell lung cancer. J Infus Chemother 5:195–196
predictors for radiation-induced esophageal injury. Int J Emami B (1996) Three-dimensional conformal radiation
Radiat Oncol Biol Phys 61:335–347 therapy in bronchogenic carcinoma. Semin Radiat Oncol
Antonadou D, Coliarakis N, Synodinou M et al (2001) 6:92–97
Randomized phase II trial of radiation treatment plus/minus Gandara DR, Chansky K, Albain KS et al (2003) Consolidation
amifostine in patients with advanced-stage lung cancer. Int J docetaxel after concurrent chemoradiotherapy in stage IIIb
Radiat Oncol Biol Phys 51:915–922 non-small cell lung cancer: phase II southwest oncology
Ball D, Bishop J, Smith J et al (1995) A phase III study of group study S9504. J Clin Oncol 21(10):2004–2010
accelerated radiotherapy with and without carboplatin in Goldstein HM, Rogers LF, Fletcher GH, Dodd GD (1975)
non-small cell lung cancer: an interim toxicity analysis of Radiological manifestations of radiation-induced injury
the first 100 patients. Int J Radiat Oncol Biol Phys 31:267– to the normal upper gastrointestinal tract. Radiology 117:
272 135–140
Belderbos J, Heemsbergen W, Hoogeman M, Pengel K, Rossi Grills IS, Yan D, Martinez AA, Vicini FA, Wong JW, Kestin
M, Lebesque J (2005) Acute esophageal toxicity in non- LL (2003) Potential for reduced toxicity and dose escalation
small cell lung cancer patients after high dose conformal in the treatment of inoperable non-small-cell lung cancer: a
radiotherapy. Radiother Oncol 75:157–164 comparison of intensity-modulated radiation therapy
Radiation Therapy-Related Toxicity: Esophagus 645

(IMRT), 3D conformal radiation, and elective nodal irradi- Sarna L, Swann S, Langer C et al (2008) Clinically meaningful
ation. Int J Radiat Oncol Biol Phys 57(3):875–890 differences in patient-reported outcomes with amifostine in
Kahn D, Zhou S, Ahn SJ et al (2005) ‘‘Anatomically-correct’’ combination with chemoradiation for locally advanced non-
dosimetric parameters may be better predictors for esoph- small-cell lung cancer: an analysis of RTOG 98–01. Int J
ageal toxicity than are traditional CT-based metrics. Int J Radiat Oncol Biol Phys 72(5):1378–1384
Radiat Oncol Biol Phys 62(3):645–651 Saunders MI, Dische S, Barrett A, Harvey A, Gibson D, Parmar
Kim TH, Cho KH, Pyo HR et al (2005) Dose-volumetric M (1997) Continuous hyperfractionated accelerated radio-
parameters of acute esophageal toxicity in patients with therapy (CHART) versus conventional radiotherapy in non-
lung cancer treated with three-dimensional conformal small cell lung cancer: a randomized multicenter trial.
radiotherapy. Int J Radiat Oncol Biol Phys 64(4):995–1002 Lancet 350:161–165
Komaki R, Lee JS, Milas L et al (2004) Effects of amifostine on Spielberger R, Stiff P, Bensinger W et al (2004) Palifermin for
acute toxicity from concurrent chemotherapy and radiother- oral mucositis after intensive therapy for hematological
apy for inoperable non-small cell lung cancer: report of a cancers. N Engl J Med 351(25):2590–2598
randomized comparative trial. Int J Radiat Oncol Biol Phys Stickle RL, Epperly MW, Klein E, Bray J, Greenberger J
58(5):1369–1377 (1999) Prevention of irradiation-induced esophagitis by
Langer C, Hsu C, Curran W et al (2001) Do elderly patients with plasmid/liposome delivery of the human manganese
locally advanced non-small cell lung cancer benefit from superoxide dismutase transgene. Radiat Oncol Invest 7:
combined modality treatment? A secondary analysis of RTOG 204–217
94–10. Int J Radiat Oncol Biol Phys 51(1 Suppl):20–21 Suit H, Goldberg S, Niemierko A et al (2003) Proton beams to
Lepke RA, Libshitz HI (1983) Radiation-induced injury of the replace photon beams in radical dose treatments. Acta
esophagus. Radiology 148:375–378 Oncol 42(8):800–808
Maguire PD, Sibley GS, Zhou SM et al (1999) Clinical and Vokes EE, Herndon JE 2nd, Crawford J et al (2002) Random-
dosimetric predictors of radiation-induced esophageal tox- ized phase II study of cisplatin with gemcitabine or
icity. Int J Radiat Oncol Biol Phys 45(1):97–103 paclitaxel or vinorelbine as induction chemotherapy fol-
McGinnis WL, Loprinzi CL, Buskirk SJ et al (1997) Placebo- lowed by concomitant chemoradiotherapy for stage IIIB
controlled trial of sucralfate for inhibiting radiation-induced non-small-cell lung cancer: cancer and leukemia group B
esophagitis. J Clin Oncol 15:1239–1243 study 9431. J Clin Oncol 20(20):4191–4198
Michalowski A, Hornsey S (1986) Assays of damage to the Werner-Wasik M, Pequignot E, Leeper D, Hauck W, Curran W
alimentary canal. Br J Cancer 53(suppl 7):1–6 (2000) Predictors of severe esophagitis include use of
Movsas B, Scott C, Langer C et al (2005) Randomized trial of concurrent chemotherapy, but not the length of irradiated
amifostine in locally advanced non-small cell lung cancer esophagus: a multivariate analysis of patients with lung
receiving chemotherapy and hyperfractionated radiation: cancer treated with non-operative therapy. Int J Radiat
radiation therapy oncology group trial 98–01. J Clin Oncol Oncol Biol Phys 48:689–696
23(10):2145–2154 Werner-Wasik M, Axelrod SA, Friedland DP et al (2002) Phase
Patel AB, Edelman MJ, Kwork Y, Krasna MJ, Suntharalingam II trial of twice weekly amifostine in patients with non-
M (2004) Predictors of acute esophagitis in patients with small cell lung cancer treated with chemotherapy. Semin
non small-cell lung carcinoma treated with concurrent Radiat Oncol 12(suppl 1):34–39
chemotherapy and hyperfractionated radiotherapy followed Werner-Wasik M, Yorke E, Deasy J, Nam J, Marks LB (2010)
by surgery. Int J Radiat Oncol Biol Phys 60:1106–1112 Radiation dose-volume effects in the esophagus. Int J Radiat
Phillips TL, Ross G (1974) Time-dose relationships in the Oncol Biol Phys 76(3 Suppl):S86–93
mouse esophagus. Radiology 113:435–440 Werner-Wasik M, Paulus R, Curran WJ Jr, Byhardt R Acute
Rodriguez N, Algara M, Foro P et al (2005) Predictors of acute esophagitis and late lung toxicity in concurrent chemor-
esophagitis in lung cancer patients treated with concurrent adiotherapy in patients with locally advanced non-small cell
three-dimentional conformal radiotherapy and chemother- lung cancer: analysis of the radiation therapy oncology
apy. Int J Radiat Oncol Biol Phys 73(3):810–817 group (RTOG) database (in press)
Brain Toxicity
C. Nieder

Contents Abstract
Since lung cancer generally has a high, stage- and
1 Introduction.............................................................. 647 histology-dependent propensity for brain metasta-
2 Pathogenesis of Radiotherapy-Induced ses, many patients receive prophylactic or thera-
Brain Toxicity .......................................................... 648 peutic radiotherapy to the brain and are therefore at
3 Acute and Subacute Radiotherapy-Induced risk for developing acute, subacute or chronic side
Brain Toxicity .......................................................... 650 effects. Certain types of systemic treatment might
also cause brain toxicity. In this chapter the
4 Delayed or Chronic Radiotherapy-Induced
Brain Toxicity .......................................................... 650 different types of toxicity, the pathogenesis and
emerging prevention and treatment strategies are
5 Toxicity Prevention Strategies ............................... 653
reviewed.
6 Treatment of Radiotherapy-Induced Brain
Toxicity...................................................................... 655
7 Aspects of Chemotherapy-Induced Brain 1 Introduction
Toxicity...................................................................... 655
References.......................................................................... 655 Prevention and treatment of brain metastases contin-
ues to be an important issue despite considerable
improvements in local and systemic therapy for lung
cancer. As long as many patients are diagnosed in
advanced stages of the disease, their risk for devel-
oping brain metastases is high. Up to 60% of patients
with small-cell lung cancer (SCLC) will be diagnosed
with brain metastases at some time during the natural
course of the disease. Therefore, prophylactic cranial
irradiation (PCI) is now recommended in the majority
of patients with SCLC. The second indication for
brain irradiation in lung cancer is palliation of
symptoms from brain metastases. Depending on the
number of lesions, their size and location and well
established prognostic factors (Nieder et al. 2009),
whole-brain radiotherapy (WBRT), open surgical
resection or stereotactic radiosurgery (SRS) may be
C. Nieder (&)
the preferred option. Another indication is adjuvant
Department of Oncology and Palliative Medicine,
Nordland Hospital, P.O. Box 1480, 8092 Bodø, Norway radiotherapy after resection of brain metastases,
e-mail: carsten.nieder@nordlandssykehuset.no which is usually administered by means of WBRT.

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 647
DOI: 10.1007/174_2011_221, Ó Springer-Verlag Berlin Heidelberg 2011
648 C. Nieder

However, adjuvant tumor bed SRS or brachtherapy is signs of diffuse demyelination develop in animals
also under investigation. This chapter will therefore 2 weeks after CNS radiotherapy. After approximately
cover the normal tissue effects of both partial brain 2 months, remyelination processes have been
radiotherapy and WBRT to the normal adult brain. observed. These early changes correspond to clinical
The issue of reduced tolerance in the immature brain symptoms such as Lhermitte’s sign and somnolence
will not be discussed, due to the fact that it is less in humans. After a variable latency period, and
relevant in the context of lung cancer. dependent on total dose, white matter necrosis may
The physical and technical developments and develop. The gray matter is less sensitive. Latency
refinements over the last decades in radiation oncol- time decreases with increasing radiation dose. The
ogy have been impressive. But still, the easiest and most important determinants of CNS tolerance are the
most effective way of avoiding side effects to the volume of normal tissue exposed, dose per fraction
normal brain is by minimizing its exposure to ionizing and total dose. Overall treatment time is less impor-
radiation. While individually-shaped, highly confor- tant. With multiple fractions per day, incomplete
mal dose distributions can be created, this does not repair needs to be taken into account, especially when
solve the problem of the presence of normal tissue the interfraction interval is less than 6 h.
within the irradiated target volume (the result of dif- When WBRT is being administered, the complete
fuse microscopic spread, which escapes current intracranial vascular system is exposed to ionizing
imaging technology). Therefore, many patients con- radiation, although at relatively modest doses, in
tinue to receive WBRT. Where a reduction of the contrast to focal treatment where only limited parts of
irradiated volume is not feasible, further progress can the blood vessels might receive a significantly higher
only be expected from efforts directed at optimizing dose. When high focal doses are combined with lower
fractionation or widening the therapeutic window doses to a large surrounding volume, tolerance
between tumor and normal tissue through modulation decreases, compared with the same focal treatment
of the patient’s responses to radiotherapy. alone.
We will discuss the pathogenesis of radiation- Significant long-term recovery has been observed
induced brain toxicity, the incidence of typical side after spinal cord radiotherapy. Although not experi-
effects, risk factors, diagnostic aspects and the role of mentally tested in the same fashion, it can be assumed
multimodal treatment concepts in the development of that the brain recovers, too. Especially with larger
side effects. Increasing evidence can be found in the intervals of at least 1–2 years and when the first
literature about the influence of cytotoxic drugs and treatment course was not too close to tolerance, re-
the general side effects of cancer treatment, such as irradiation is now considered as a realistic option.
anemia, on the normal brain. Finally, pre-clinical and Experimental data from fractionated radiotherapy of
clinical data on the prevention and treatment of side rhesus monkeys suggest a recovery of up to 75% of
effects will be reviewed. the initial damage within 2–3 years (Ang et al. 2001).
The past few years have witnessed a significant
improvement as far as techniques are concerned in
2 Pathogenesis of Radiotherapy- cellular and molecular biology, resulting, for exam-
Induced Brain Toxicity ple, in a description of more and more radiobiologi-
cally-relevant cellular pathways. Better methods for
Early evaluations of radiotherapy-induced central the identification of stem and progenitor cells have
nervous system (CNS) toxicity date back to over been developed. This progress has led to a better
70 years ago. It is not the aim of this chapter to dis- understanding of tissue responses to ionizing radia-
cuss these historical data, which have been summa- tion. Obviously, radiation-induced reactions of the
rized in previous reviews, for example, by van der CNS are not limited to reproductive or mitotic cell
Kogel (1986). When appropriate, data from spinal death in mature parenchymal and vascular cell pop-
cord radiotherapy will be included in the current ulations. Apoptosis, induced by sphingomyelinase-
chapter because of the similarity of radiation-induced mediated release of ceramide, has been described as
changes in the brain and the spinal cord. In brief, an early reaction in endothelial cells within the
previous experimental studies have indicated that irradiated CNS (Pena et al. 2000), as well as in
Brain Toxicity 649

oligodendrocytes (Larocca et al. 1997). Besides cell apoptosis (Hisahara et al. 1997; Akassoglou et al.
death, a large number of alterations in gene expres- 1998; Gu et al. 1999), the cytokine in vitro prevents
sion, transcription factor activation and functional the differentiation of O-2A progenitor cells into oli-
changes in basically every cell type examined may godendrocytes. Thus, compensation for radiation-
develop (Raju et al. 2000). Current models of radio- induced cell loss can be impaired. TNF-a is also
therapy-induced brain alterations include a cascade of known to damage endothelial cells, leading to
complex and dynamic interactions between paren- increased vascular permeability. TNF-a and IL-1
chymal cells (oligodendrocytes, astrocytes, microg- induce the expression of intercellular adhesion mol-
lia), stem and progenitor cells and vascular ecule-1 (ICAM-1) in oligodendrocytes and micro-
endothelial cells (Tofilon and Fike 2000; Fike et al. vascular endothelial cells (Satoh et al. 1991; Wong
2009). The latent time preceding the clinical mani- and Dorovini 1992). Increased levels of ICAM-1
festation of damage is viewed as an active phase, mRNA were detectable after midbrain irradiation
where cytokines and growth factors play important with 2 Gy (Hong et al. 1995). Results of localized
roles in intra- and intercellular communication single-fraction treatment with 20 Gy confirm the
(Nieder et al. 2007). Clinically recognized phenom- presence of an early inflammatory response, an
ena, such as intellectual decline, memory loss, leth- increased numbers of leukocytes, increased vascular
argy, dysphoria, dementia and ataxia, also suggest the permeability, altered integrity of endothelial tight
possible involvement of neurons in radiotherapy- junctions and increased cell adhesion (Yuan et al.
induced CNS reactions. In vitro studies have dem- 2003). The exact role of such cytokines and mediators
onstrated that neurons may undergo apoptosis after after radiotherapy with conventional fraction sizes is
radiotherapy (Gobbel et al. 1998). Fractionated brain not well understood yet; clearly, the cellular and
irradiation inhibited the formation of new neurons in molecular events during the latent phase require fur-
the dentate gyrus of the hippocampus in rats (Madsen ther research.
et al. 2003). Animals with blocked neurogenesis Studies of boron neutron capture therapy (BNCT)
performed poorer in short-term memory tests that are support the view that vascular damage is one of the
related to hippocampal function. The deficit in neu- crucial components of radiotherapy-induced CNS
rogenesis is based on both the reduced proliferative toxicity. The choice of boron compounds that are
capacity of progenitor cells and alterations in the unable to cross the blood–brain barrier allows a lar-
microenvironment that regulates progenitor cell fate gely-selective irradiation of the vessel walls with
(disruption of the microvascular angiogenesis, acti- BNCT. Nevertheless as with conventional non-
vation of microglia) (Monje et al. 2002, 2007). selective radiotherapy methods, spinal cord lesions
CNS radiotherapy induces production of inflam- (with a similar histological appearance) have been
matory cytokines, such as tumor necrosis factor a induced. Latency time also is comparable between
(TNF-a) and interleukin-1 (IL-1), by microglia and damage induced by BNCT and conventional radio-
astrocytes (Chiang and McBride 1991; Hayakawa therapy (Morris et al. 1996). Additional evidence has
et al. 1997). IL-1 release leads, via autocrine mech- been provided by histological examinations of rat
anisms, to further activation and proliferation of these brains after radiotherapy with 22.5 or 25 Gy, resulting
glia cells. As shown in vivo, this cascade results in the in reduced numbers of blood vessels and endothelial
development of astrogliosis (Chiang et al. 1993). cells before manifestation of necrosis (Calvo et al.
Already 2 h after single-fraction radiotherapy to the 1988). These changes are accompanied by hyperper-
midbrain of mice (25 Gy), TNF-a and IL-1 mRNA meability, resulting in perivascular edema and con-
levels have been shown to increase (Hong et al. secutive ischemic damage (Hopewell and van der
1995). After 24 h, the levels start returning to normal. Kogel 1999). Microvascular networks, consisting of
Experimental rat brain irradiation has also been arterioles, capillaries and venoles, which impact the
shown to induce apoptosis, which, in turn, appears to delivery of oxygen and nutrients to tissues and organs,
result in an increase in the number of microglial cells are the most radiosensitive parts of the vascular sys-
participating in phagocytotic reactions. Besides the tem (Roth et al. 1999). Common therapeutic doses
cytotoxic effects of TNF-a on oligodendrocytes, for of ionizing radiation lead to functional and, later,
example, through induction of caspase-mediated to structural vascular damage, such as increased
650 C. Nieder

permeability and changes in shape and diameter, as endothelial growth factor (VEGF) as a result of
well as in fibrous proliferation, ultimately resulting in impaired perfusion and hypoxia signaling. Obviously,
reduced perfusion. These changes develop earlier in the clinically observed latent phase is characterized
small versus large vessels. After lower doses, struc- by persistent and increasing oxidative stress and
tural changes are hardly ever seen. After WBRT in active responses to this factor. The extreme sensitivity
rats (five fractions of 4 Gy) alterations in vessel of the myelin membrane to oxidative damage explains
configuration, either density or diameter, were not the preference of radiotherapy-induced lesions for
detected (Mildenberger et al. 1990). Interestingly, a white matter.
localized significant increase of microglia was found
after 6 months, possibly as a result of the loss of
axons in the striatal white matter. The pattern was 3 Acute and Subacute Radiotherapy-
suggestive of vascular insufficiency in this region, Induced Brain Toxicity
which was being perfused by only few small vessels.
Electron microscopy in rats 15 days after the end of As stated in the previous paragraph, acute and suba-
conventional fractionated WBRT (40 Gy) showed cute radiotherapy-induced brain toxicity can develop
increased vascular permeability without structural within hours from the start of treatment even if low
changes of the blood–brain barrier or astrocytes doses are given. It is usually characterized by
(Cicciarello et al. 1996). A follow-up examination increased vascular permeability, edema and demye-
after 90 days revealed ultrastructural changes of the lination manifesting as headache, nausea, somnolence
microvasculature and the neuropil, as well as astro- or lethargy. However, it has been characterized as a
cytes with perivascular edema. temporary, self-limiting reaction, which responds to
Another study (partial brain irradiation with 40 or corticosteroid treatment (Schultheiss et al. 1995).
60 Gy, or WBRT with 25 Gy in rats) showed a 15% Subacute reactions may develop 2–6 months after
reduction in the number of endothelial cells 24 h to WBRT, resulting, for example, in lethargy, reduced
4 weeks after radiotherapy. A further reduction was vigilance and impaired cognitive performance. Most
seen with even longer intervals (Ljubimova et al. likely, such symptoms are related to a second phase of
1991). Kamiryo et al. showed how the latency to transient demyelination and blood–brain barrier dis-
development of vascular damage after SRS to the turbance. Treatment with corticosteroids again is
parietal cortex of rat brain with a 4 mm collimator likely to improve the patient’s condition. With SRS,
decreases from 12 months to 3 weeks with an acute reactions are rare. They include symptomatic
increase in radiation dose from 50 to 75 Gy or edema, seizures and nausea and vomiting, especially
120 Gy (Kamiryo et al. 1996). The amount of vessel when doses [3.75 Gy are given to the area postrema.
dilation, increased permeability, thickening of the Antiemetics, corticosteroids and anticonvulsant drugs
vessel wall, vessel occlusion and necrosis also may be used to treat these symptoms. Temporary
increased with dose. In a different model of rat brain blood–brain disturbance may result in increased
irradiation, time and dose-dependent vascular altera- contrast enhancement in computed tomography (CT)
tions were also seen (dilation, wall thickening, reac- or MRI during the first few months after SRS. These
tive hypertrophy of neighboring astrocytes) before the changes should not be misinterpreted as tumor pro-
development of white matter necrosis (Hopewell et al. gression. Usually they resolve with longer follow-up.
1989). Rubin et al. performed comprehensive mag-
netic resonance imaging (MRI) and histological
examinations of rat brains after 2–24 weeks following 4 Delayed or Chronic Radiotherapy-
high dose, single-fraction irradiation with 60 Gy Induced Brain Toxicity
(Rubin et al. 1994). After 2 weeks, a significant
increase in blood–brain permeability was observed. Sustaining toxicity that may impair the patient’s
Partial recovery occurred after 8–12 weeks, followed lifestyle significantly can be observed even several
by pronounced deterioration after 24 weeks, when the years after radiotherapy in the form of radionecrosis
first sites of necrosis developed. Spinal cord data and cognitive dysfunction associated with leukoen-
suggest an increase in the release of vascular cephalopathy. Necrosis develops for the most part
Brain Toxicity 651

after 1–3 years (Keime-Guibert et al. 1998). Symp-


toms of radionecrosis depend on localization and are
comparable to tumor-related symptoms before treat-
ment (focal neurologic deficits and seizures, speech
disturbance, signs of increased intracranial pressure).
CT and T1- and T2-weighted anatomic MRI are
unable to firmly discriminate between hypometabolic
necrosis and tumor relapse. Dynamic susceptibility
contrast-enhanced MRI, magnetic resonance spec-
troscopy (MRS) and functional imaging by means of
positron emission tomography (PET) and 201Tl-single
photon emission computed tomography (SPECT) can
provide useful additional information (Munley et al.
2001; Nakajima et al. 2009). Eventually, in some
cases, only histopathological examination of resection
specimens can establish the diagnosis. The typical
finding is coagulation necrosis in the white matter,
with a largely normal appearance of the cortex.
Fibrinoid necrosis and hyalinous wall thickening of
blood vessels are commonly observed. The risk of
radionecrosis amounts to approximately 5% within
5 years (ED5/5) after conventional fractionated partial
brain radiotherapy (one third of the brain) with 60 Gy
or WBRT with 45 Gy. The dose–response curves are
quite steep. Thus the risk increases to 10% within
5 years when a partial brain dose of 65 Gy is applied
(according to data from the randomized U.S. inter-
group low-grade glioma trial) and 50% when 75 Gy is
given. Irradiated volume, dose per fraction and total
dose are the most important risk factors (Lawrence
et al. 2010). Recent series reported radionecrosis after
SRS of brain metastases in 1–6% of cases, probably
dependent on brain region and vascular supply. Fig. 1 Initial CT scans (top) before whole-brain radiotherapy
Commonly prescribed doses are in the range of 15– (30 Gy in 10 fractions of 3 Gy) in a 66 year old man with
20 Gy, depending on volume, technique of SRS and multiple brain metastases from small cell lung cancer (no
previous prophylactic brain irradiation). The lower CT scan
use of additional WBRT. The risk increases when was taken 2 years later and shows moderate brain atrophy and
more than 10 cm3 of the normal brain receives more white matter changes. In addition periventricular calcifications
than 10 Gy. The optic apparatus should not receive (white arrow) can be seen. The patient had developed overt
more than 8 Gy (Mayo et al. 2010). Varlotto et al. neurocognitive decline interfering with activities of daily living
reported the results of SRS in 137 patients with brain
metastases who had a minimum follow-up of 1 year B2 cm3 and 16% for those with larger lesions. Age
after SRS (Varlotto et al. 2003). The median marginal and additional use of WBRT did not influence the
tumor dose was 16 Gy. NSCLC was the underlying complication rate. Therapeutic intervention with
primary tumor in 77 patients. Eleven patients devel- corticosteroids or anticoagulants is sometimes suc-
oped serious side effects, such as visual loss, hemor- cessful (Glantz et al. 1994). Often, surgical resection
rhage and persistent steroid-dependent edema or is the only way to effectively improve the symptoms.
necrosis necessitating surgical intervention. The Diffuse white matter changes are frequently observed
actuarial incidence of such adverse events was 4% in imaging studies (Figs. 1 and 2). Fluid-attenuated
after 5 years for patients with brain metastases inversion recovery (FLAIR) and diffusion-weighted
652 C. Nieder

the temporal lobe, the hippocampal formation plays a


central role in short-term memory and learning. These
functions are related to the activity of neural stem
cells. The hippocampal granule cell layer undergoes
continuous renewal and restructuring. Radiotherapy
can affect this sensitive cell layer leading to impaired
function without overt pathological changes. Our own
retrospective data from 49 patients who had received
WBRT with a median dose of 30 Gy showed that
33% of patients developed mild to moderate clinical
symptoms of brain toxicity (in one case, RTOG/EO-
RTC grade III, median follow-up 10 months, median
dose per fraction 3 Gy) (Nieder et al. 1999). This
resulted in a Karnofsky-performance status decline in
10 patients (20%). None of the PCI patients belonged
to this subgroup. CT showed increasing brain atrophy
and bilateral periventricular hypodensity in most
patients (Fig. 1). The actuarial risk of brain atrophy
was 84% after 2 years. Median time to development
of this side effect was 11 months. Patients with pre-
existing brain atrophy had a higher risk of further
shrinkage of the brain parenchyma than those with
normal baseline status. White matter changes were
observed in 85% of surviving patients. Radiologic
abnormalities did not correlate with the rate of clin-
ical symptoms. Previous studies described such cor-
relations for patients treated with PCI and
chemotherapy for SCLC (Laukkanen et al. 1988;
Johnson et al. 1990). Whether or not clinical symp-
toms, quality of life and radiologic abnormalities
correlate, might depend on variables such as length of
follow-up, methods of assessment and severity of
clinical symptoms.
When evaluating radiotherapy-induced cognitive
Fig. 2 Initial T2-weighted MRI scan before stereotactic radi- impairment, it is important to consider reference
osurgery (gamma knife, peripheral dose 20 Gy) in a 67 years values from the normal population. A Canadian Study
old woman with solitary brain metastasis from non-small-cell
lung cancer. The lower MRI scan was taken 2.8 years later and
of Health and Aging with 9,008 randomly selected
shows focal white matter changes (white arrow). No clinical men and women 65 years or older showed cognitive
late toxicity was apparent impairment 5 years after baseline examination in 9%
and dementia in an additional 6% (Laurin et al. 2001).
MRI may improve visualization of white matter Decline was significantly associated with reduced
abnormalities. These abnormalities are not necessarily physical activity. There is increasing evidence that
associated with clinical symptoms but often present partial brain radiotherapy alone rarely causes signifi-
after fractionated doses of C30 Gy. Neuropsycho- cant neurocognitive decline (Torres et al. 2003;
logical sequelae typically manifest within 4 years of Duchstein et al. 2003). After WBRT, neuropsycho-
radiotherapy. Psychometric findings suggest greater logical tests in 29 patients showed no significant
vulnerability of white matter and subcortical struc- decrease (Penitzka et al. 2002). In this study, patients
tures, resulting in reduced processing speed, height- with SCLC were significantly below average before
ened distractibility and memory impairment. Within PCI, but did not deteriorate further. Patients with
Brain Toxicity 653

fewer cycles of preceding chemotherapy performed anterior and medial cerebral arteries) have occasion-
better before PCI. These results are in accordance ally been described, mostly in patients irradiated at a
with earlier prospective findings where 97% of younger age. Endocrine dysfunction resulting from
patients with SCLC had cognitive dysfunction prior to damage to the pituitary gland or the hypothalamic
PCI (Komaki et al. 1995). Six to twenty months later, region can result in hypothyroidism, amenorrhea, etc.
no further deterioration was identified. Some authors Hearing loss is very uncommon after doses typically
found indications for increased toxicity when fraction prescribed for lung cancer metastases.
size exceeded 2 Gy (Herskovic and Orton 1986; Importantly, all types of iatrogenic neurotoxicity
Twijnstra et al. 1987; De Angelis et al. 1989). Also can only be diagnosed after comprehensive evaluation
the large intergroup study that used PCI with 10 excluding other causes, for example brain metastases,
fractions of 2.5 Gy in patients with SCLC reported leptomeningeal spread, infections, cerebral infarction
mild deterioration of intellectual and memory func- and hemorrhage. In addition, systemic metabolic
tion (Le Pechoux et al. 2011). The largest study in disorders (hypercalcaemia, hepatic failure, diabetes,
patients with NSCLC was reported by Sun et al. changes in osmolality etc.), alcoholic cerebellar
(2011). Randomization between PCI (30 Gy in 15 degeneration, Wernicke-Korsakoff syndrome and
fractions of 2 Gy) and observation was done in a paraneoplastic disorders (for example, limbic
group of 356 patients. PCI was associated with a encephalitis, chorea, cerebellar degeneration and
significant decline in memory at 1 year (Hopkins Lambert-Eaton myasthenic syndrome in SCLC) must
Verbal Learning Test; recall deterioration in 26 vs. be considered. Besides physical and neurologic
7% and delayed recall deterioration in 32 vs. 5%). examination, blood tests, EEG and cerebrospinal fluid
Three months after PCI, but not at later time points, diagnostic are indicated. In addition to imaging
there was also a significant deterioration in Mini- studies—for example, myelography, CT, MRI and
Mental-Status examination (36 vs. 21%). In patients functional imaging—factors such as time interval
who developed brain metastases, WBRT has also between radiotherapy and diagnosis, dose per frac-
been shown to interfere with recall functions as tion, number of fractions per day, total dose and
measured by the Hopkins Verbal Learning Test (64% location of the treatment fields need to be considered.
in patients treated with WBRT plus SRS as compared In the era of multimodal treatment regimens, injury
to 20% in those with SRS alone after 4 months, i.e. in should not be attributed solely to one modality.
the subacute phase) (Chang et al. 2009). However, Therefore, interdisciplinary evaluation integrating the
such decline might also precede progression of brain radiobiological knowledge of radiation oncologists is
metastases. In patients with impaired neurocognition mandatory when radiotherapy-induced neurotoxicity
before WBRT who respond to this treatment is being considered.
improvements might be observed. Ongoing trials
examine WBRT with hippocampal avoidance in order
to better preserve neurocognitive functions. 5 Toxicity Prevention Strategies
Neurocognitive dysfunction was reported to sta-
bilize spontaneously (van de Pol et al. 1997; Arm- At present, pharmacologic or biologic prevention is
strong et al. 2002) or to progress over time (Johnson not clinically established despite intriguing pre-
et al. 1990, Regine et al. 2001). In extreme cases, liminary data, e.g. from a non-randomized trial of
subcortical dementia may result which often is asso- SRS of arteriovenous malformations, where patients
ciated with gait disturbance and incontinence. Due to treated with gamma linolenic [omega-6] acid had less
the lack of effective treatment, most patients with this permanent complications than those who did not
severe complication die after several months or a few receive this medication (Sims and Plowman 2001).
years. Histopathologic findings include diffuse spon- Therefore, the most effective way of toxicity pre-
giosis and demyelination, as well as disseminated vention is a reduction of fraction size and normal
miliary necrosis. tissue volume, the latter, for example, by means of
Further late complications in terms of stenosis of several high-precision techniques. However, rational
blood vessels and moyamoya syndrome (multiple, experimental interventions based on the pathogenetic
diffuse, progressive infarctions due to occlusion of the models reviewed in this chapter have been studied or
654 C. Nieder

are currently under investigation. The clinical effec- time of radiation treatment, and they offer new strat-
tiveness of these putative prevention strategies has yet egies of toxicity prevention.
to be established. The prophylactic use of dexa- The growing body of evidence linking radiation-
methasone 24 and 1 h before radiation exposure induced brain injury with oxidative stress and/or
reduced the expression of TNF-a, IL-1 and ICAM-1 inflammation has provided the rationale for applying
(Hong et al. 1995). In vitro, corticosteroids influence proven anti-inflammatory-based interventions to pre-
the function of microglial cells and inhibit their pro- vent radiation-induced cognitive impairment. Recent
liferation (Tanaka et al. 1997). The hyperpermeability studies have tested drugs that can either attenuate
of blood vessels could be reduced at all time points inflammation or reduce chronic oxidative stress,
after irradiation by application of rh-MnSOD (man- namely peroxisome proliferator-activated (PPAR)
ganese superoxide dismutase), suggesting that free agonists and renin angiotensin system (RAS) block-
oxygen radicals could be involved in the dysfunction ers. Given the putative role of oxidative stress/
of microvessels. Fike et al. reported that i.v. injection inflammation in radiation-induced brain injury, Zhao
of a-difluoromethylornithine (DMFO), a polyamine et al. (2007) tested the hypothesis that administration
synthesis inhibitor, starting 2 days before and of the PPARc agonist, pioglitazone (Pio), would
continuing for 14 days after 125I brachytherapy mitigate the severity of radiation-induced cognitive
reduced the volume of radionecrosis in irradiated dog impairment. Indeed, administering Pio to young adult
brain (Fike et al. 1994). Kondziolka et al. irradiated rats starting prior to, during, and for 4 or 54 weeks
rats with implanted cerebral C6 glioma by SRS, either after the completion of fractionated WBRT, prevented
with or without i.v. administration of U-74389G, a cognitive impairment measured 52 weeks post-irra-
21-aminosteroid which is largely selective for endo- diation. Robbins et al. (2009) hypothesized that
thelium (Kondziolka et al. 1999). The compound blocking the brain RAS with the AT1RA, L-158,809,
reduced the development of peritumoral edema and would prevent or ameliorate radiation-induced cog-
radiation-induced vascular changes in the parts of the nitive impairment. As hypothesized, administering
brain that were within the region of the steep dose L-158,809 before, during, and for 28 or 54 weeks
gradient outside the target volume. No tumor pro- after fractionated WBRT prevented or ameliorated
tection was observed. In general, normal tissue cognitive impairment observed 26 and 52 weeks
selectivity of prevention approaches is an important postirradiation. Moreover, giving L-158,809 before,
issue. Protecting tumor cells against the effects of during, and for only 5 weeks postirradiation amelio-
radiation can counteract the effort of improving the rated the significant cognitive impairment seen
therapeutic ratio. 26 weeks postirradiation.
More recent data suggest the possible role of cer- Transplantation of stem cells or stimulation of the
tain growth factors with antiapoptotic effects that also endogenous stem cell compartment by growth factor
influence the proliferation of stem cells, neurogenesis application might also offer exciting prospects. In
and angiogenesis. Pena et al. showed that i.v. injec- principle, mature functional cells can be generated by
tions of basic fibroblast growth factor (FGF-2) 5 min proliferation and differentiation from stem and pro-
before, immediately after and 1 h after total body genitor cells or by recovery and repair of damage in
irradiation in mice (1–20 or 50 Gy) significantly already existing cells, which then continue to survive.
reduced the number of apoptotic vascular and glial IGF-1 has been found to influence the restoration of
cells in the CNS (Pena et al. 2000). Spinal cord neurogenesis in the adult and aging hippocampus
experiments suggest that other growth factors, such as (Lichtenwalner et al. 2001) and might thus offer inter-
platelet-derived growth factor (PDGF) and insulin- esting prospects. In another recently published exper-
like growth factor-1 (IGF-1) can increase the long- iment, athymic nude rats subjected to head irradiation
term radiation tolerance by approximately 5–10% were transplanted 2 days afterward with human
(Andratschke et al. 2005). Whether these effects result embryonic stem cells (hESC) into the hippocampal
primarily from protection of the vascular system or formation and analyzed for stem cell survival, differ-
from more widespread action is presently not known. entiation, and cognitive function (Acharya et al. 2009).
The experiments, however, demonstrate that delayed Animals receiving hESC transplantation exhibited
toxicity can be prevented by early intervention at the superior performance on a hippocampal-dependent
Brain Toxicity 655

cognitive task 4 months post-irradiation, compared to 5-fluorouracil (5-FU), methotrexate and paclitaxel.
their irradiated counterparts that did not receive hESCs. White matter hyperintensity from vasogenic edema can
Significant stem cell survival was found at 1 and clinically result in headache, somnolence and seizures.
4 months postirradiation. Symptoms can be reversed when the drugs are dis-
continued. Cerebellar toxicity of 5-FU is rare and
mostly found in patients with a deficiency of dihydro-
6 Treatment of Radiotherapy- pyrimidine dehydrogenase. Cisplatin is able to induce
Induced Brain Toxicity cerebral edema and cortical blindness, as reviewed by
(Sloan et al. 2003; Rajeswaran et al. 2008). Mild to
Despite improvements in biologic understanding of moderate, typically transient neurocognitive impair-
CNS reactions, treatment options unfortunately are ment can develop after systemic chemotherapy, for
still limited. It is of course important to exclude other example with paclitaxel (Ahles and Saykin 2001;
causes of CNS dysfunction, to correct any metabolic Herbst et al. 2002) and cisplatin/etoposide (Whitney
abnormality and to optimize the treatment of et al. 2008). Chronic encephalopathy also can result
endocrinological dysfunction, depression and other from chemo- or radiochemotherapy (Keime-Guibert
comorbid conditions. A few case reports have et al. 1998).
described successful treatment of late CNS toxicity by
hyperbaric oxygen treatment (HBO). For example,
one out of seven patients with cognitive impairment at
least 1.5 years after radiotherapy improved after References
30 sessions of HBO (Hulshof et al. 2002). In contrast,
HBO during radiotherapy can cause radiosensitiza- Acharya MM, Christie LA, Lan ML et al (2009) Rescue of
radiation-induced cognitive impairment through cranial
tion. Patients with leukencephalopathy and moderate transplantation of human embryonic stem cells. Proc Natl
hydrocephalus (diagnosed by intracranial pressure Acad Sci USA 106:19150–19155
monitoring) may profit from ventriculoperitoneal Ahles TA, Saykin A (2001) Cognitive effects of standard-dose
shunt insertion (Perrini et al. 2002). Quality of life can chemotherapy in patients with cancer. Cancer Invest
19:812–820
be improved by supportive measures (cognitive Akassoglou K, Bauer J, Kassiotis G et al (1998) Oligodendro-
training, rehabilitation, special education etc.) and cyte apoptosis and primary demyelination by local TNF/
possibly by methylphenidate medication (Meyers p55TNF receptor signaling in the central nervous system of
et al. 1998). Memantine and donepezil are also under transgenic mice: models for multiple sclerosis with primary
oligodendrogliopathy. Am J Pathol 153:801–813
investigation. For radionecrosis, therapeutic inter- Andratschke N, Nieder C, Price RE, Rivera B, Ang KK (2005)
vention with bevacizumab might offer new prospects Potential role of growth factors in diminishing radiation
(Levin et al. 2011). Previously, surgical resection therapy neural tissue injury. Semin Oncol 32(Suppl 3):S67–
often was the only way to effectively improve the S70
Ang KK, Jiang GL, Feng Y, Stephens LC, Tucker SL, Price RE
symptoms. As suggested from a recent double-blind (2001) Extent and kinetics of recovery of occult spinal cord
placebo controlled trial intravenous bevacizumab injury. Int J Radiat Oncol Biol Phys 50:1013–1020
given every 3 weeks might improve both clinical and Armstrong CL, Hunter JV, Ledakis GE et al (2002) Late
radiological symptoms. cognitive and radiographic changes related to radiotherapy:
initial prospective findings. Neurology 59:40–48
Calvo W, Hopewell JW, Reinhold HS, Yeung TK (1988) Time-
and dose-related changes in the white matter of the rat brain
7 Aspects of Chemotherapy-Induced after single doses of X-rays. Br J Biol 61:1043–1052
Brain Toxicity Chang EL, Wefel JS, Hess KR et al (2009) Neurocognition in
patients with brain metastases treated with radiosurgery or
radiosurgery plus whole-brain irradiation: a randomised
Chemotherapy can cause a variety of brain injuries. Most controlled trial. Lancet Oncol 10:1037–1044
of these changes are temporary and reversible. Some- Chiang CS, McBride WH (1991) Radiation enhances tumor
times the symptoms are secondary to hyponatremia or necrosis factor alpha production by murine brain cells.
Brain Res 566:265–269
hypomagnesemia. Posterior reversible encephalopathy Chiang CS, McBride WH, Withers HR (1993) Radiation-
syndrome can develop after systemic administration induced astrocytic and microglial responses in mouse brain.
of cytotoxic drugs, including gemcitabine, cisplatin, Radiother Oncol 29:60–68
656 C. Nieder

Cicciarello R, D’Avella D, Gagliardi ME et al (1996) Time- Kamiryo T, Kassell NF, Thai QA, Lopes MB, Lee KS, Steiner
related ultrastructural changes in an experimental model of L (1996) Histological changes in the normal rat brain after
whole brain irradiation. Neurosurgery 38:772–779 gamma irradiation. Acta Neurochir 138:451–459
De Angelis LM, Delattre JY, Posner JB (1989) Radiation- Keime-Guibert F, Napolitano M, Delattre JY (1998) Neuro-
induced dementia in patients cured of brain metastases. logical complications of radiotherapy and chemotherapy.
Neurology 39:789–796 J Neurol 245:695–708
Duchstein S, Gademann G, Peters B (2003) Early and late Komaki R, Meyers CA, Shin DM et al (1995) Evaluation of
effects of local high dose radiotherapy of the brain on cognitive function in patients with limited small cell lung
memory and attention [Article in German]. Strahlenther cancer prior to and shortly following prophylactic cranial
Onkol 179:441–451 irradiation. Int J Radiat Oncol Biol Phys 33:179–182
Fike JR, Gobbel GT, Marton LJ, Seilhan TM (1994) Radiation Kondziolka D, Mori Y, Martinez AJ, McLaughlin MR,
brain injury is reduced by the polyamine inhibitor a- Flickinger JC, Lunsford LD (1999) Beneficial effects of
difluoromethylornithine. Radiat Res 138:99–106 the radioprotectant 21-aminosteroid U-74389G in a radio-
Fike JR, Rosi S, Limoli CL (2009) Neural precursor cells and surgery rat malignant glioma model. Int J Radiat Oncol Biol
central nervous system radiation sensitivity. Semin Radiat Phys 44:179–184
Oncol 19:122–132 Larocca JN, Farooq M, Norton WT (1997) Induction of
Glantz MJ, Burger PC, Friedman AH, Radtke RA, Massey EW, oligodendrocyte apoptosis by C2-ceramide. Neurochem Res
Schold SC (1994) Treatment of radiation-induced nervous 22:529–534
system injury with heparin and warfarin. Neurology Laukkanen E, Klanoff H, Allan B, Graeb D, Murray N (1988)
44:2020–2027 The role of prophylactic brain irradiation in limited stage
Gobbel GT, Bellinzona M, Vogt AR, Gupta N, Fike JR, Chan small cell lung cancer: clinical, neuropsychological, and CT
PH (1998) Response of postmitotic neurons to x-irradiation: sequelae. Int J Radiat Oncol Biol Phys 14:1109–1117
implications for the role of DNA damage in neuronal Laurin D, Verreault R, Lindsay J, Rockwood K (2001) Physical
apoptosis. J Neurosci 18:147–155 activity and risk of cognitive impairment and dementia in
Gu C, Casaccia-Bonnefil P, Srinivasan A, Chao MV (1999) elderly persons. Arch Neurol 58:498–504
Oligodendrocyte apoptosis mediated by caspase activation. Lawrence YR, Li XA, El Naqa I, Hahn CA, Marks LB,
J Neurosci 19:3043–3049 Merchant TE, Dicker AP (2010) Radiation dose-volume
Hayakawa K, Borchardt PE, Sakuma S, Ijichi A, Niibe H, effects in the brain. Int J Radiat Oncol Biol Phys 76:S20–
Tofilon PJ (1997) Microglial cytokine gene induction after S27
irradiation is affected by morphologic differentiation. Radiat Le Pechoux C, Laplanche A, Faivre-Finn C et al (2011)
Med 15:405–410 Clinical neurological outcome and quality of life among
Herbst RS, Madden TL, Tran HT et al (2002) Safety and patients with limited small-cell cancer treated with two
pharmacokinetic effects of TNP-470, an angiogenesis different doses of prophylactic cranial irradiation in the
inhibitor, combined with paclitaxel in patients with solid intergroup phase III trial (PCI 99-01, EORT 22003-08004,
tumors. J Clin Oncol 20:4440–4447 RTOG 0212 and IFCT 99-01). Ann Oncol [epub ahead of
Herskovic AM, Orton CG (1986) Elective brain irradiation for print]
small cell anaplastic lung cancer. Int J Radiat Oncol Biol Levin VA, Bidaut L, Hou P, Kumar AJ, Wefel JS, Bekele BN,
Phys 12:427–429 Prabhu S, Loghin M,Gilbert MR, Jackson EF (2011)
Hisahara S, Shoji S, Okano H, Miura M (1997) ICE/CED3 Randomized double-blind placebo-controlled trial of-
family executes oligodendrocyte apoptosis by tumor necro- bevacizumab therapy for radiation necrosis of the central
sis factor. J Neurochem 69:10–20 nervous system. Int JRadiat Oncol Biol Phys. 79(5):1487–
Hong JH, Chiang CS, Campbell IL, Sun JR, Withers HR, 1495
McBride WH (1995) Induction of acute phase gene Lichtenwalner RJ, Forbes ME, Bennett SA, Lynch CD,
expression by brain irradiation. Int J Radiat Oncol Biol Riddle DR (2001) Intracerebroventricular infusion of
Phys 33:619–626 insulin-like growth factor-1 ameliorates the age-related
Hopewell JW, van der Kogel AJ (1999) Pathophysiological decline in hippocampal neurogenesis. Neuroscience 107:
mechanisms leading to the development of late radiation- 603–613
induced damage to the central nervous system. Front Radiat Ljubimova NV, Levitman MK, Plotnikova ED, Eidus LK
Ther Oncol 33:265–275 (1991) Endothelial cell population dynamics in rat brain
Hopewell JW, Calvo W, Campling D, Reinhold HS, Rezvani after local irradiation. Br J Radiol 64:934–940
M, Yeung TK (1989) Effects of radiation on the microvas- Madsen TM, Kristjansen PE, Bolwig TG, Wortwein G (2003)
culature. Front Radiat Ther Oncol 23:85–95 Arrested neuronal proliferation and impaired hippocampal
Hulshof MC, Stark NM, van der Kleij A, Sminia P, Smeding function following fractionated irradiation in the adult rat.
HM, Gonzalez D (2002) Hyperbaric oxygen therapy for Neuroscience 119:635–642
cognitive disorders after irradiation of the brain. Strahlen- Mayo C, Martel MK, Marks LB, Flickinger J, Nam J,
ther Onkol 178:192–198 Kirkpatrick J (2010) Radiation dose-volume effects of optic
Johnson BE, Patronas N, Hayes W et al (1990) Neurologic, nerves and chiasm. Int J Radiat Oncol Biol Phys 76:S28–
computed cranial tomographic, and magnetic resonance S35
imaging abnormalities in patients with small-cell lung Meyers CA, Weitzner MA, Valentine AD (1998) Methylphe-
cancer: further follow-up of 6- to 13-year survivors. J Clin nidate therapy improves cognition, mood and function of
Oncol 8:48–56 brain tumor patients. J Clin Oncol 16:2522–2527
Brain Toxicity 657

Mildenberger M, Beach TG, McGeer EG, Ludgate CM (1990) radiotherapy: an analysis from RTOG study 91–04. Int J
An animal model of prophylactic cranial irradiation: Radiat Oncol Biol Phys 51:711–717
histologic effects at acute, early and delayed stages. Int J Robbins ME, Payne V, Tommasi E et al (2009) The AT1
Radiat Oncol Biol Phys 18:1051–1060 receptor antagonist, L-158, 809, prevents or ameliorates
Monje ML, Mizumatsu S, Fike JR, Palmer TD (2002) fractionated whole-brain irradiation-induced cognitive
Irradiation induces neural precursor-cell dysfunction. Nat- impairment. Int J Radiat Oncol Biol Phys 73:499–505
ure Med 8:928–930 Roth NM, Sontag MR, Kiani MF (1999) Early effects of
Monje ML, Vogel H, Masek M et al (2007) Impaired human ionizing radiation on the microvascular networks in normal
hippocampal neurogenesis after treatment for central ner- tissue. Radiat Res 151:270–277
vous system malignancies. Ann Neurol 62:515–520 Rubin P, Gash DM, Hansen JT, Nelson DF, Williams JP (1994)
Morris GM, Coderre JA, Bywaters A (1996) Boron neutron Disruption of the blood-brain barrier as the primary effect of
capture irradiation of the rat spinal cord: histopathological CNS irradiation. Radiother Oncol 31:51–60
evidence of a vascular-mediated pathogenesis. Radiat Res Satoh J, Kastrukoff LF, Kim SU (1991) Cytokine-induced
146:313–320 expression of intercellular adhesion molecule-1 (ICAM1) in
Munley MT, Marks LB, Hardenbergh PH, Bentel GC (2001) cultured human oligodendrocytes and astrocytes. J Neuro-
Functional imaging of normal tissues with nuclear medi- pathol Exp Neurol 50:215–226
cine: applications in radiotherapy. Semin Radiat Oncol Schultheiss TE, Kun LE, Ang KK, Stephens LC (1995)
11:28–36 Radiation response of the central nervous system. Int J
Nakajima T, Kumabe T, Kanamori M et al (2009) Differential Radiat Oncol Biol Phys 31:1093–1112
diagnosis between radiation necrosis and glioma progres- Sims EC, Plowman PN (2001) Stereotactic radiosurgery XII.
sion using sequential proton magnetic resonance spectros- Large AVM and the failure of the radiation response
copy and methionine positron emission tomography. Neurol modifier gamma linolenic acid to improve the therapeutic
Med Chir (Tokyo) 49:394–401 ratio. Br J Neurosurg 15:28–34
Nieder C, Leicht A, Motaref B, Nestle U, Niewald M, Schnabel Sloan AE, Arnold SM, St. Clair WH, Regine WF (2003) Brain
K (1999) Late radiation toxicity after whole-brain radio- injury: current management and investigations. Semin
therapy: the influence of antiepileptic drugs. Am J Clin Radiat Oncol 13:309–321
Oncol 22:573–579 Sun A, Bae K, Gore EM et al (2011) Phase III trial of
Nieder C, Andratschke N, Astner ST (2007) Experimental prophylactic cranial irradiation compared with observation
concepts for toxicity prevention and tissue restoration after in patients with locally advanced non-small-cell lung
central nervous system irradiation. Radiat Oncol 2:23 cancer: neurocognitive and quality-of-life analysis. J Clin
Nieder C, Bremnes RM, Andratschke NH (2009) Prognostic Oncol 29:279–286
scores in patients with brain metastases from non-small cell Tanaka J, Fujita H, Matsuda S, Toku K, Sakanaka M, Maeda N
lung cancer. J Thorac Oncol 4:1337–1341 (1997) Glucocorticoid- and mineralocorticoid receptors in
Pena LA, Fuks Z, Kolesnick RN (2000) Radiation-induced microglial cells: the two receptors mediate differential
apoptosis of endothelial cells in the murine central nervous effects of corticosteroids. Glia 20:23–37
system: protection by fibroblast growth factor and sphingo- Tofilon PJ, Fike JR (2000) The radioresponse of the central
myelinase deficiency. Cancer Res 60:321–327 nervous system: a dynamic process. Radiat Res 153:357–
Penitzka S, Steinvorth S, Sehlleier S, Fuss M, Wannenmacher 370
M, Wenz F (2002) Assessment of cognitive function after Torres IJ, Mundt AJ, Sweeney PJ, Castillo M, Macdonald RL
preventive and therapeutic whole brain irradiation using (2003) A longitudinal neuropsychological study of partial
neuropsychological testing [Article in German]. Strahlen- brain radiation in adults with brain tumors. Neurology
ther Onkol 178:252–258 60:1113–1118
Perrini P, Scollato A, Cioffi F, Conti R, Di Lorenzo N (2002) Twijnstra A, Boon PJ, Lormans ACM, Ten Velde GPM (1987)
Radiation leukoencephalopathy associated with moderate Neurotoxicity of prophylactic cranial irradiation in patients
hydrocephalus: intracranial pressure monitoring and with small cell carcinoma of the lung. Eur J Cancer Clin
results of ventriculoperitoneal shunting. Neurol Sci 23: Oncol 23:983–986
237–241 Van de Pol M, Ten Velde GP, Wilmink JT, Volovics A,
Rajeswaran A, Trojan A, Burnand B, Giannelli M (2008) Twijnstra A (1997) Efficacy and safety of prophylactic
Efficacy and side effects of cisplatin- and carboplatin-based cranial irradiation in patients with small cell lung cancer.
doublet chemotherapeutic regimens versus non-platinum- J Neurooncol 35:153–160
based doublet chemotherapeutic regimens as first line Van der Kogel AJ (1986) Radiation-induced damage in the
treatment of metastatic non-small cell lung carcinoma: a central nervous system: an interpretation of target cell
systematic review of randomized controlled trials. Lung responses. Br J Cancer 53(Suppl 7):207–217
Cancer 59:1–11 Varlotto JM, Flickinger JC, Niranjan A, Bhatnagar AK,
Raju U, Gumin GJ, Tofilon PJ (2000) Radiation-induced Kondziolka D, Lunsford LD (2003) Analysis of tumor
transcription factor activation in the rat cerebral cortex. Int J control and toxicity in patients who have survived at least
Radiat Biol 76:1045–1053 one year after radiosurgery for brain metastases. Int J Radiat
Regine WF, Scott C, Murray K, Curran W (2001) Neurocog- Oncol Biol Phys 57:452–464
nitive outcome in brain metastases patients treated with Whitney KA, Lysaker PH, Steiner AR, Hook JN, Estes DD,
accelerated-fractionation vs. accelerated-hyperfractionated Hanna NH (2008) Is ‘‘chemobrain’’ a transient state? A
658 C. Nieder

prospective pilot study among persons with non-small cell leukocyte adhesion in the rat blood–brain barrier: mod-
lung cancer. J Support Oncol 6:313–321 ulation with anti-ICAM-1 antibodies. Brain Res 969:59–
Wong D, Dorovini ZK (1992) Up-regulation of intercellular 69
adhesion molecule-1 (ICAM-1) expression in primary cul- Zhao W, Payne V, Tommasi E et al (2007) Administration of
tures of human brain microvessel endothelial cells by the peroxisomal proliferator-activated receptor (PPAR)c
cytokines and lipopolysaccharide. J Neuroimmunol 39:11–21 agonist pioglitazone during fractionated brain irradiation
Yuan H, Gaber MW, McColgan T, Naimark MD, Kiani MF, prevents radiation-induced cognitive impairment. Int J
Merchant TE (2003) Radiation-induced permeability and Radiat Oncol Biol Phys 67:6–9
Quality of Life Outcomes in Radiotherapy
of Lung Cancer
M. Salim Siddiqui, Farzan Siddiqui, and Benjamin Movsas

Contents Abstract
Lung cancer is one of the most common cancers in
1 Introduction.............................................................. 661 the world. There has been an increased interest in
1.1 What is Quality of Life............................................. 662 quality of life (QOL) as a clinically meaningful
1.2 How is QOL Measured ............................................. 662
1.3 How is QOL Analyzed.............................................. 666
endpoint in clinical trials for lung cancer. Patient-
1.4 QOL Studies in Lung Cancer ................................... 668 reported outcome (PRO) measurements have
allowed oncologists to better understand the
2 Conclusions and Future Directions ....................... 670
impact of cancer therapies on the physical, emo-
References.......................................................................... 671 tional and social well-being of their patients. This
chapter provides a review of the clinical relevance,
measurement, analysis and challenges of quality of
life in lung cancer trials. A summary of salient
quality-of-life studies, particularly involving radi-
ation treatment for lung cancer, is included.

1 Introduction

Over the past two decades, quality of life (QOL) has


not only grown in interest, as evidenced by the
increasing QOL literature, but also has become a
well-accepted endpoint in clinical trials for cancer, in
general, and lung cancer, specifically. As focus on
improving physical, emotional, and social well-being
has increased, QOL outcome measurements have
allowed oncologists to better understand the impact of
various cancer therapies on QOL and to make
M. Salim Siddiqui  B. Movsas (&) recommendations to improve the overall QOL of the
Department of Radiation Oncology, patient. Such improvements are even more important
Henry Ford Hospital, 2799 W Grand Boulevard, when one considers the poor prognosis in the majority
Detroit, MI 48202, USA
e-mail: bmovsas1@hfhs.org of lung cancer patients. In many cases, treatments for
lung cancer may be associated with toxicities which
F. Siddiqui
Department of Radiation Oncology, Ohio State University, could adversely impact QOL. In such settings,
300 W. 10th Avenue, Columbus, OH 43210, USA assessment of QOL endpoints may help to identify

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 661
DOI: 10.1007/174_2011_259, Ó Springer-Verlag Berlin Heidelberg 2011
662 M. Salim Siddiqui et al.

significant differences in such treatment-related tox- only its definition and quantification, but also its
icities, while the traditional endpoints may be equiv- scientific study in the realm of clinical trials. Broadly,
ocal. QOL studies afford knowledge to accurately according to the World Health Organization (WHO),
communicate potential treatment-related changes in ‘‘Quality of life is defined as individuals’ perceptions
functional and emotional well-being to future of their position in life in the context of the culture
patients. As a result, efforts can be directed to prevent and value system where they live, and in relation to
or mitigate such toxicities or changes in well-being. their goals, expectations, standards and concerns. It is
Since 1985, the United States Food and Drug a broad ranging concept incorporating in a complex
Administration (FDA) recognized the importance of way a person’s physical health, psychological state,
QOL by including the requirement for a favorable level of independence, social relationships, personal
effect on survival and/or QOL in the approval of new beliefs and relationship to salient features of the
anti-cancer drugs (Johnson and Temple 1985). Drugs environment’’ (WHO 1996). The WHO defined health
such as gemcitabine, irinotecan, vinorelbine, oxyco- as ‘‘a state of complete physical, mental and social
done, mitoxantrone, and erythropoietin have been well-being and not merely the absence of disease’’.
approved, at least in part, based on QOL endpoints
(Leitgeb et al. 1994; Burris et al. 1997). Similarly, the
National Cancer Institute (NCI) has also emphasized 1.2 How is QOL Measured
the importance of improving QOL in cancer patients
as exemplified in the mission statement of the Divi- There are several major considerations to address
sion of Cancer Treatment of the NCI which states that regarding the measurement of QOL in clinical studies.
‘‘research aimed at improving survival and QOL for First, self-reporting by the patient remains the gold
persons with cancer is of the highest priority to the standard for measuring an individual’s QOL.
Cancer Therapy Evaluation Program’’ (NCI, C. T. C. Attempts to rate QOL by physicians, other medical
G. P 1988). The NCI convenes QOL conferences to personnel, or even family members, have been found
develop recommendations for including QOL to be significantly different from patient reports
assessments in clinical trials (Cella and Tulsky 1990; (Slevin et al. 1988; Watkins-Bruner et al. 1995). The
Strain 1990). importance of such patient-reported outcomes (PROs)
Given the continued growth of QOL outcomes in has been recognized by both the NCI and the FDA.
cancer, and particularly in lung cancer, this chapter A PRO is defined as any report of the status of a
will provide an overview of the significance, meth- patient’s health condition provided directly by the
odology, analysis, limitations and challenges of QOL patient, without interpretation of the patient’s
studies. The chapter will include relevant examples of response by anyone else. Examples of PROs include
QOL studies in lung cancer. QOL, treatment preferences, symptoms, and satis-
faction with care. However, PROs are not yet
standardized in adverse event reporting. Currently,
1.1 What is Quality of Life physicians tend to underestimate the frequency and
severity of patients’ symptoms when reporting
The inherent subjective and abstract nature of the adverse events utilizing health-care provider instru-
term ‘‘Quality of Life’’ provides a considerable ments (e.g. NCI Common Terminology Criteria for
challenge to define this construct. In addition, the Adverse Events, or CTCAE). As a result, the current
notion of QOL spans a broad range of an individual’s reporting structure likely under-represents the true
feelings, beliefs and perceptions of life. As a result, treatment-related toxicity burden in clinical trials. To
QOL remains a fluid concept, influenced by various address this discrepancy between physician-reported
social, physical, financial, cultural and emotional outcomes and PROs, particularly for adverse events,
factors. The challenges to objectively quantify this the NCI has initiated the PRO-CTCAE project to
construct are further compounded by the inter- and create patient-reported versions of those symptom
intra- patient cross-cultural and temporal variability criteria.
of QOL (Leplege and Hunt 1997). However, appli- The next major consideration centers on whether
cation of this construct in the clinic necessitates not QOL should be measured in all clinical trials.
Quality of Life Outcomes in Radiotherapy of Lung Cancer 663

Finite financial, data management, and human an instrument can be assessed in terms of content
resources, limit the measurement of QOL outcomes in (i.e. does it address the relevant issues), construct (i.e.
clinical trials. Moreover, QOL outcomes may serve does it indeed measure what it claims to measure),
limited function in certain settings, such as phase I and clinical focus (i.e. does it differentiate patient
and II trials, which usually aim to answer questions groups with differing clinical and/or sociodemo-
about maximally tolerated doses and preliminary graphic features). Simply put, a valid test must
efficacy data for new treatments. Inclusion of QOL appropriately focus on the intended target and then
measures in such trials would not only create undue measure the outcome that it claims to measure.
patient burden, but also afford limited return. Rather, Responsiveness refers to the ability of an instrument
Phase III randomized trials, which allow for the to detect changes in an outcome over time within a
careful design and implementation of a well-formu- particular individual or group (Husted et al. 2000).
lated QOL hypothesis, provide a more meaningful Similar to responsiveness, sensitivity is the ability of
clinical arena to measure such outcomes. Gotay the instrument to measure true changes or differences
(2004) suggested the following guidelines for inclu- in outcome. Both the responsiveness and sensitivity of
sion of QOL endpoints in a Phase III trial: (1) If QOL an instrument may be affected by ‘‘ceiling’’ or ‘‘floor’’
is considered to be the primary endpoint, such as a effects when outcome scores from patient response
study comparing two palliative regimens. (2) If the rest at the limits of the possible score range. In such
treatments are expected to be equivalent in terms of circumstances, the instrument would not allow for
efficacy (e.g. survival), such that one treatment would measurement of any positive or negative movement
be considered preferable if it is associated with a beyond the ‘‘ceiling’’ or ‘‘floor’’, respectively.
relative QOL advantage. (3) If the advantage of one Regardless of such effects, a sensitive and responsive
arm in terms of outcome may be real, but nevertheless instrument should measure stable outcomes when
offset by increased toxicity and deterioration in QOL. there is no change in outcome. After reviewing the
(4) If the treatments differ in short-term efficacy, but reliability, validity, sensitivity, and responsiveness of
the overall failure rate is high (Gotay 2004). Simi- an instrument, the actual task of selecting the
larly, Moinpour et al. (1989) proposed the following ‘‘optimal’’ QOL instrument remains. Primarily, this
guidelines: (1) the disease site is associated with a selection should focus on the QOL instrument that the
poor prognosis, (2) different treatment modalities are investigator(s) believe is most suited to answer the
being compared, (3) different treatment intensities hypothesis of the study and that is most relevant for
and/or duration are being compared, and (4) expected the population studied in the clinical trial. Four con-
survival is assumed equivalent, but the QOL is siderations have been suggested when selecting the
expected to be different between the two regimens. ‘‘best’’ QOL instrument: (1) the underlying hypothe-
The third critical consideration is the selection of sis or purpose of the trial, (2) the patient population,
the most appropriate QOL instrument. In reviewing, (3) the treatments and their toxicity profiles, and (4)
testing and validating any QOL instrument, particular the degree of resources available (Gelber and Gelber
attention must be placed on the reliability, validity, 1995).
sensitivity, and responsiveness to change over time of
the instrument (Testa and Simonson 1996). The FDA 1.2.1 QOL Instruments
issued a guidance document on PROs which encodes QOL instruments can be categorized by their focus
these standards and specifies that PRO measures and/or by their targeted clinical population. Generic
should demonstrate reliability, validity,and sensitivity QOL instruments have the broadest focus and are
to score changes, and have appropriate recall periods designed to assess the general health status of
(FDA 2009). The reliability quantifies the freedom individuals with a broad range of diagnoses. How-
from random error of the instrument. In other words, a ever, the inherent breath of these questionnaires may
reliable instrument reports values that remain con- result in their inability to account for clinical issues in
sistent with repeated measurement under constant site-, condition, or treatment-specific measures.
conditions. Generally, an instrument reliability of Examples of such instruments include the Sickness
0.70 or higher is considered acceptable for clinical Impact Profile (SIP) (Bergner et al. 1976, 1981),
trials (Nunnaly and Bernstein 1994). The validity of the Medical Outcomes Study (MOS) (Stewart and
664 M. Salim Siddiqui et al.

Ware 1992), and the Medical Outcomes Study 36 multi-item scales: five functional scales (physical,
item Short Form Health Survey (MOS SF-36) (Ware role, cognitive, emotional, and social); three symp-
and Sherbourne 1992; McHorney et al. 1993, 1994). tom scales (fatigue, pain, and nausea and vomiting);
The Sickness Impact Profile (SIP) is a self-adminis- and additional global health and quality-of-life
tered 136 item questionnaire that assesses physical scales. In addition to the EORTC QLQ-C30, the
and psycho-social domains and 12 categories (sleep QLQ-LC13 has been developed and validated for
and rest, eating, work, home management, recreation lung cancer (Bergman et al. 1994). The QLQ-LC13
and pastimes, ambulation, mobility, body care and is a 13 item lung cancer specific questionnaire that
movement, social interaction, alertness behavior, consists of one three-question subscale (dyspnea)
emotional behavior and communication) (Bergner and 10 symptom items (cough, hemoptysis, sore
et al. 1976, 1981). The Medical Outcomes Study is a mouth, dysphagia, peripheral neuropathy, alopecia,
116 item questionnaire that assesses physical health pain in chest, pain in shoulder, pain and pain med-
(i.e. physical functioning, satisfaction with physical ication). The score of each item ranges from 0 to
ability, mobility, pain effects, pain severity, role 100 with better functioning indicated by a higher
limitations due to physical health), mental health value on the functional scales and increased symp-
(i.e. psycho-social distress, anxiety and depression, toms indicated by a higher score on the symptom
psychological well-being, cognitive functioning,and items. The QLQ-LC13 questionnaire has also been
role limitations due to emotional problems), and validated for lung cancer patients (Nicklasson and
general health (i.e. energy/fatigue, sleep problems, Bergman 2007).
psycho-physiologic symptoms, social functioning, The FACT-G instrument consists of a 27-item
role functioning) (Stewart and Ware 1992). A trun- questionnaire measuring physical well-being (7-items),
cated version of the MOS, the MOS 36-item Short social/family well-being (7-items), emotional well-
Form survey (MOS SF-36) is widely used for routine being (6-items) and functional well-being (7-items).
outcome studies in adult patients (Ware and Sher- Each item can be scored from 0 to 4 with a range of 0–28
bourne 1992; McHorney et al. 1993, 1994). Higher for physical, social/family and functional well-being
scores on this form correlate with a better QOL. and 0–24 for emotional well-being and a total score
Other QOL instruments have been developed that range of 0–108. This oncology questionnaire has been
combine generic and specific measurements into one validated and is also available in over 50 languages
instrument. Such instruments assess patients with a (Cella et al. 1993). In addition to the FACT-G, the
specific clinical condition through a set of common FACT-L has been developed and validated for lung
core questions plus a set of clinical condition-specific cancer (Cella et al. 1995). The FACT-L is a 9 item lung
questions. Examples of this type of instrument in cancer specific questionnaire that assesses dyspnea,
oncology are the European Organization for Research cough, weight loss, chest pain, alopecia, appetite loss,
and Treatment of Cancer (EORTC), Quality of Life and smoking. In contrast to the EORTC QLQ-LC13,
Questionnaire Core 30 (QLQ-C30) (Aaronson et al. the FACT-L does not assess hemoptysis, sore mouth,
1993) and the Functional Assessment of Cancer dysphagia, or peripheral neuropathy. Of note, the
Therapy-General (FACT-G) (Cella et al. 1993). For FACT-TOI (trial outcome index) combines the physical
lung cancer patients, in addition to generic and well-being and functional well-being domains of the
condition-specific QOL instruments, site-specific FACT-G with the FACT-L subscale. The FACT-TOI
instruments are sometimes used, such as the Lung has also been validated in lung cancer patients (Cella
Cancer Symptom Scale (LCSS) (Hollen et al. 1993, et al. 2002).
1994a, 1994b). The Lung Cancer Symptom Scale (LCSS) consists
The EORTC QLQ-C30 is an oncology instrument of a 9 item patient-reported questionnaire and an
containing 30 items with scores from 0 (not at all) to 4 optional 6 item observer questionnaire (Hollen et al.
(very much) for items 1–28 and from 1 (very poor) to 1993, 1994a, 1994b). The patient-reported question-
7 (excellent) for items 29 and 30. The instrument has naire consists of six lung cancer symptoms
been validated (Aaronson et al. 1993) and has been (i.e. appetite loss, fatigue, cough, dyspnea, hemopty-
translated in over 50 languages. The EORTC QLQ- sis, and pain) and three general items (i.e. symptom-
C30 assesses various elements of QOL through nine atic distress, activity status, and overall QOL).
Quality of Life Outcomes in Radiotherapy of Lung Cancer 665

The optional observer portion consists of a question- statistical power, thus affecting the generalizability of
naire assessing the same six lung cancer symptoms as the results to the larger population. Low compliance
in the patient-reported portion. The score for each may result in either ‘‘missing items’’ or ‘‘missing
item is summed into a total score between 0 and 100. forms’’ (Fayers et al. 1998), also known as ‘‘item non-
A higher score correlates with a better QOL (Hollen response’’ and ‘‘unit non-response’’, respectively
et al. 1994a). (Troxel et al. 1998).
In the case of ‘‘missing items’’, the patient does not
1.2.2 Frequency and Timing of QOL respond to some questions on the QOL questionnaire.
Measurements This lack of response may be due to a number of
After selection of the most appropriate QOL instru- causes, including, but not limited to, the patient was
ment, attention must be turned to establishing the too ill or distressed, or forgot, or was unable to
optimal timing and frequency of administration of the understand the questions (Ganz et al. 1989; Curran
instrument. Such administration will vary between et al. 1998; Langendijk et al. 2000b; Aaronson and
clinical trials and will be influenced by the hypothesis Fayers 2002). Such missing data results in informa-
and the treatment protocol of the clinical trial, the tive censoring, which may underestimate or overes-
natural course of the disease, and the anticipated side- timate the QOL of patients. For example, consider the
effects of treatment (Osoba 1996). Regardless of these ‘‘healthy survivor’’ effect, which results when patients
factors, every clinical trial with QOL endpoints who are sicker or not doing well on treatment do not
should perform a pre-treatment QOL assessment. provide complete responses on QOL questionnaires.
Such an assessment not only establishes a baseline An association between such ‘‘nonignorable’’ missing
QOL, which is essential for QOL analysis, but also data and patients’ status has been suggested
has prognostic significance for lung and other cancers (Moinpour et al. 2000). While most patients, who
(Coates et al. 1997; Dancey et al. 1997; Langendijk agree to enroll on QOL studies, understand the benefit
et al. 2000a; Blazeby et al. 2001; de Graeff et al. their contribution makes for medical research and
2001; Montazeri et al. 2001; Fang et al. 2004; Efficace future patients, they may not appreciate that such
and Bottomley 2005; Movsas et al. 2009). The base- benefit can only be fully realized if complete
line assessment also determines differences in pre- responses are provided. Of course, patients should be
treatment QOL in treatment groups and individuals informed of their choice not to answer any question(s)
and allows comparison to post-treatment QOL. Dur- that make them feel uncomfortable.
ing the course of the clinical trial, the frequency of The case of ‘‘missing forms’’, which is more
QOL data collection must balance the cost of data serious than ‘‘missing items’’, can be further catego-
collection/management and patient burden with the rized as: (1) intermittent missing forms, (2) dropouts
benefit of the additional QOL data. The frequency of from the study, or (3) patients entering the study late
collection can be directed by a time-based (e.g. at a (Curran et al. 1998). Possible explanations for such
set time after randomization) or an event-based (e.g. missing forms may be: (1) administrative failure to
after a specific treatment cycle) approach. Typically, distribute the questionnaire, (2) patient considered the
QOL assessments at the completion of treatment and questionnaire a violation of privacy, (3) patient
post-completion (i.e. a few months later) are helpful. thought the questionnaire time consuming, (4) patient
Such measures afford not only the study of treatment refusal or withdrawal, (5) patient felt too ill, and/or
side-effects, but also knowledge regarding the course (6) disease progression (Curran et al. 1998). Cer-
and nature of recovery from those side-effects. Such tainly, clinical investigators can anticipate and
knowledge can be particularly useful in making address some of these explanations through adequate
clinical decisions and recommendations. infrastructure and logistical support for data collec-
tion and careful and thoughtful reassurance and
1.2.3 Compliance encouragement of patients.
Low compliance, or missing data, remains the most For QOL to be a key component of a clinical trial,
important challenge in QOL trials. Loss of such data high-patient compliance and minimal data loss need
may affect the integrity of the QOL study, by to be a goal. In addition, quality control measures
increasing the tendency toward bias and decreasing should ensure standard procedures for administering
666 M. Salim Siddiqui et al.

and reviewing the QOL questionnaires. Often such 1.3 How is QOL Analyzed
responsibilities are performed by data managers or
trained research nurses. Centralizing such responsi- The challenge in the statistical analysis of QOL data
bilities and establishing quality control measures may arises from the inherent complexity of the data and
result in high-compliance rates (Sadura et al. 1992; the desire to present the results in a simple and
Langendijk et al. 2000a). comprehensible manner. Ideally, the results are not
Beyond traditional centralized QOL data collec- only statistically significant, but also clinically rele-
tion by mail or telephone, real-time tracking soft- vant. The concern of ‘‘overanalysis’’ plagues such
ware may also be utilized to maintain records and large and complex databases with fears of spurious
alert personnel regarding missing data. Recently, statistically significant relationships. As a result,
such QOL data tracking has been piloted by the various statistical methods have been proposed to
Radiation Therapy Oncology Group (RTOG) in a understand and interpret QOL data. A review article
study (RTOG 0828) attempting to reduce missing by Wyrwich et al. (2005) and the Clinical Signifi-
QOL data. The Health Insurance Portability and cance Consensus Meeting Group discusses these
Accountability Act (HIPPA)-complaint software for issues more comprehensively. For the purpose of this
internet-based reporting can track particular ques- chapter, some of these methods and relevant literature
tions not routinely being answered, alert medical are briefly reviewed.
research personnel about missing items and ask
patients if they would like to answer the missing 1.3.1 Anchor-Based and Distribution-Based
questions or not. This allows for automatic e-mail Analysis
reminders for patients to complete QOL question- The anchor-based method of analysis correlates the
naires. Moreover, this may allow patients to log-in scores on the HRQOL instrument used in a study (i.e.
from outside the clinic (i.e. home, office, etc.) to the target instrument) with another independent
complete questionnaires, not only minimizing time measure (i.e. the anchor). This method can be further
spent in the clinic, but also allowing less disruption categorized by the nature of the anchor as individual-
of their daily lives. Ideally, assuming broad-scale focused (i.e. single-anchor) or population-focused
internet access, such real-time data tracking affords a (i.e. multiple-anchor) (Guyatt et al. 2002). The indi-
feedback loop to assess the level of completion rates vidual-focused method attempts to address the ques-
between questionnaires, determines questions con- tion of whether small changes in QOL are clinically
sistently not answered, and thus, may help minimize relevant. By defining a threshold between important
missing data. and trivial change, this method introduces the concept
Regardless of the various strategies employed to of minimum important difference (MID), which is
maximize patient compliance and minimize data ‘‘the smallest difference in the score in the domain of
loss, missing data are common in QOL studies. As a interest that patients perceive as important, either
result, any QOL study should document the number beneficial or harmful, and which would lead the cli-
of missing questionnaires throughout the course of nician to consider a change in the patient’s manage-
the study and provide an explanation for missing ment’’(Guyatt et al. 2002). Osoba et al. (1998) used a
data. Advanced statistical methods have been pro- series of EORTC QLQ-C30 questionnaires to inves-
posed in literature to deal with missing data, tigate QOL and a subjective significance question-
including index function models, propensity scores, naire (SSQ) to investigate the MID. The SSQ allowed
instrumental variables, and simple and multiple assessment of ‘‘subjectively significant’’ perception of
imputation methods (Rubin 1997; Curran et al. 1998; change since the last time the patient completed the
Fairclough et al. 1998; Fayers et al. 1998; Hahn QLQ C30. In the SSQ, the patient scored their per-
et al. 1998; Troxel et al. 1998; Zee 1998; Qian et al. ception of change with a seven-category scale ranging
2000; Little and Rubin 2002; Ribaudo and from ‘‘much worse’’ to ‘‘no change’’ to ‘‘much
Thompson 2002; Sales et al. 2004). Comprehensive better’’. They found that if the mean change in scale
discussion of these statistical methods is beyond the scores on the QLQ C30 was zero, the patients per-
scope of this chapter. ceived ‘‘no change’’ in their condition. However, if
Quality of Life Outcomes in Radiotherapy of Lung Cancer 667

the mean change in scale scores was from 5 to 10 An inherent limitation of the distribution-based
points, patients indicated their condition was ‘‘a little’’ method arises from the difference in the measure of
better/worse. Similarly, for mean change in scale variability between studies as a result of patient group
scores of 10–20 and [20, patients perceived that and population heterogeneity. Wyrwich and
change in their condition was ‘‘moderate’’ and ‘‘very colleagues have suggested that the standard error of
much’’ better/worse, respectively. In a similar man- measurement may afford a solution to this inherent
ner, Cella and colleagues used the FACT-L ques- limitation and may correlate with the MID (Wyrwich
tionnaire to investigate QOL and clinically et al. 1999a, 1999b, 2002; Wyrwich 2004).
meaningful change (CMC) to determine the MID
(Cella et al. 2002). They found that a 2–3 point 1.3.2 Clinical Relevance
change in the Lung Cancer Subscale and a 5–7 point Regardless of the method of analysis, the most
change in the Trial Outcome Index were clinically important result of any QOL study, for both the
meaningful (Cella et al. 2002). These examples physician and the patient, is the clinical relevance of
highlight how the use of the MID approach affords the the QOL outcome. Effectively, does the result con-
determination of clinically meaningful changes in the tribute in a clinically meaningful manner to treatment
scores of QOL instruments for patients. As the name benefit and/or risk? In the case of baseline QOL,
implies, the population-focused, or the multiple- multiple studies have shown the independent
anchor, method correlates target measure with mul- prognostic significance of this outcome for survival
tiple anchors, instead of defining thresholds, such as and/or loco-regional control (Coates et al. 1997;
the MID. Please refer to Ware and Keller for an Dancey et al. 1997; Langendijk et al. 2000b; Blazeby
example of the population-focused method (Ware et al. 2001; de Graeff et al. 2001; Montazeri et al.
et al. 1996). 2001; Fang et al. 2004; Efficace and Bottomley 2005;
In the distribution-based method, interpretation of Movsas et al. 2009). Indeed, Movsas et al. (2009)
the data centers on the relationship between the reported that the baseline QOL score was indepen-
magnitude of change and some measure of variability dently predicted for 5-year overall survival in patients
in results (Guyatt et al. 2002) or the underlying dis- with Stage III NSCLC treated with chemoradiation.
tribution of the results (Movsas 2003). The magnitude Given this relationship, the question still remains,
of change analyzed may occur in the score of an how mitigating patients’ symptoms, and thus
individual patient or patient group (i.e. treatment and improving patients’ QOL would affect this prognostic
control groups). Because, some degree of variability, significance.
either between-patient or within-patient, exists in As for other QOL outcomes, the clinical impact
QOL data sets and, for that matter, most large data must be understood in the context of the overall
sets, this approach may appear advantageous over the treatment regimen. For example, in the case of
anchor-based methods. Cohen has proposed a criteria palliative regimens, while traditional endpoints may
that a change of 0.2 SD correlates with a ‘‘small’’ be equivocal, QOL endpoints may demonstrate clin-
change in a measure, while changes of 0.5 and 0.8 SD ically meaningful differences. For example, in a
correlate with ‘‘moderate’’ and ‘‘large’’ changes in the double-blinded, palliative study of advanced endo-
measure, respectively (Cohen 1988; Guyatt et al. crine-insensitive tumors, Beller et al. (1997) ran-
2002). While some have found this approach arbitrary domized 240 patients to either 12 weeks of high- or
(Guyatt et al. 2002) and others have found the MID low-dose megesteral acetate (MA) versus a placebo.
from their studies to rest in the 0.2–0.5 SD range Nutritional status (weight, skinfold thickness and
(Osoba et al. 1998; Norman et al. 2003) have found midarm circumference) and QOL (using six linear
that the threshold for perception of change in QOL analogue self-assessment [LASA] scales) were
measure is 0.5 SD. They performed a review 38 assessed at randomization and after 4, 8 and
studies that determined MID. In all but six studies, the 12 weeks. While the traditional ‘‘objective’’ nutri-
MID values were approximately 0.5 SD. They sug- tional status endpoints did not demonstrate a statisti-
gested that this may result from the limit of humans to cally significant difference, the QOL endpoints
discriminate on a scale, which is approximately one demonstrated significant improvement in appetite,
part in seven (Miller 1956), or approximately 0.5 SD. mood, and overall QOL in patients receiving MA.
668 M. Salim Siddiqui et al.

Even in studies with an expected difference in sur- fractionated palliative treatment was recommended.
vival between two study arms, QOL outcome can help Surprisingly, in contrast to these results, in another
weigh the risk/benefits of the various treatments. The randomized trial (Macbeth et al. 1996) by the Medical
challenge remains when QOL outcomes from a nec- Research Council Lung Cancer Working Party,
essary treatment indicate an unavoidable detriment to Macbeth and co-workers compared the F2 regimen
the QOL of the patient. In such a setting, beyond versus 39 Gy in 13 fractions (i.e. F13 regimen) in 509
providing a needed awareness of the treatment chal- patients with inoperable NSCLC and found that while
lenges for the patient, such QOL results necessitate the F2 regimen had more rapid palliation and less
the development of interventions aimed at mitigating dysphagia, the F13 regimen had longer survival.
treatment-induced QOL detriment. Similarly, Erridge et al. (2005) randomized 149
advanced lung cancer patients to palliative radiation
with either 10 Gy in one fraction or 30 Gy in 10
1.4 QOL Studies in Lung Cancer fractions. Although both treatment regimens met the
pre-determined 20% difference in the number of
The number of studies investigating QOL outcomes patients achieving improvement in the total symptom
in patients with small-cell lung cancer (SCLC) and score and had equivalent treatment toxicity, signifi-
non-small-cell lung cancer (NSCLC) has continued to cantly more patients experienced complete resolution
grow. Much of this interest stemmed from the poor and palliation of chest pain and dyspnea with the
prognosis and palliative intent of many treatment fractionated regimen. In another randomized trial,
regimens for patients with advanced lung cancer. Bezjak et al. (2002) compared 10 Gy in 1 fraction
However, beyond palliative regimens, as definitive versus 20 Gy in 5 fractions for palliation of thoracic
treatment regimens continue to increase, QOL studies symptoms in 230 patients with inoperable lung can-
may help clinically weigh the risks and benefits of cer. Interestingly, as judged by patient daily diary
treatment regimens with similar traditional outcomes. scores, no significant difference was found between
A comprehensive summary of lung cancer studies that the two treatment arms at 1-month post RT; however,
have assessed QOL endpoints is beyond the scope of QOL scores measured using the Lung Cancer
this chapter. However, a review of some salient QOL Symptom Scale indicated that the fractionated RT
studies, particularly involving radiation treatment for group (i.e. five fractions group) had significant
lung cancer is included. improvement in symptoms related to lung cancer,
Multiple randomized trials have been guided by pain, ability to conduct normal activities, and better
QOL assessments in investigating palliative irradia- global quality of life. Moreover, patients in this
tion regimens for patients with inoperable NSCLC. treatment group survived on average 2 months longer
Bleehen et al. (1991) compared 30 Gy in ten fractions with no significant difference in treatment-related
or 27 Gy in six fractions versus 17 Gy in two frac- toxicity. Recently, Temel et al. (2010) assessed the
tions for 369 patients with inoperable NSCLC. QOL impact of early palliative care on PROs and end-
assessment was not significantly different between of-life care among ambulatory patients with newly
treatment arms. As the traditional endpoint of survival diagnosed metastatic NSCLC. Using the FACT-L
was also not significantly different between these scale and the Hospital Anxiety and Depression scale,
treatment arms, the two fraction regimen (i.e. F2 they evaluated QOL and mood, respectively, in 151
regimen) was recommended. Similarly, Sundstrom patients with metastatic NSCLC at baseline and at
et al. (2004) studied three palliative irradiation regi- 12 weeks. Patients were randomized to early pallia-
mens (i.e. 17 Gy/2 fractions versus 42 Gy/15 frac- tive care or standard care. They found that patients in
tions versus 50 Gy/25 fractions) for 421 patients with the early palliative care arm not only had significant
Stage III or IV NSCLC. Using the EORTC QLQ-C30 improvement in both QOL and mood, but also had
and QLQ-LC13 instruments, they assessed QOL at longer median survival (11.6 months vs. 8.9 months,
baseline and at 2, 6, 14, 22, 30, 38, 46, and 54 weeks p = 0.02) despite receiving less therapy (Temel et al.
from the start of radiation therapy. No significant 2010).
differences were observed in median survival, symp- Beyond QOL studies for palliative radiation
tom relief, or QOL. As a result, short-term hypo treatments in lung cancer patients, prospective
Quality of Life Outcomes in Radiotherapy of Lung Cancer 669

longitudinal studies in such patients undergoing rad- QOL changes during and after radiation treatment
ical or curative radiation treatments have also been using the EORTC QLQ-30 and QLQ-LC13. In con-
completed. Langendijk et al. (2001) assessed QOL trast to conventional study above (Langendijk et al.
measures in 164 patients with NSCLC who received 2000a), in the stereotactic study (van der Voort et al.
radical radiotherapy (i.e. 60 Gy in 24 fractions). In 2010), QOL scores were maintained and emotional
addition, the study also investigated the association functioning significantly improved, while local
between level of symptom relief and objective tumor recurrence and toxicity were low and survival was
response and radiation-induced pulmonary changes. acceptable. As more evidence continues to suggest
This study was one of the first to follow the dynamics that stereotactic radiotherapy should play an increas-
of QOL changes during and after radiation treatment ing role for managing medically inoperable Stage I
using the EORTC QLQ-30 and QLQ-LC13. Radical NSCLC patients, Louie et al. (2010) developed a
radiotherapy offered not only good palliation of Markov model to compare the quality-adjusted life
respiratory symptoms, but also improved QOL in a expectancy and overall survival in medically operable
good proportion of patients. Specifically, the QOL Stage I NSCLC undergoing conventional standard of
response rates were excellent for hemoptysis (83%); care surgery versus stereotactic radiotherapy. Their
good for arm/shoulder pain (63%), chest wall pain model revealed that stereotactic radiotherapy may
(68%), and appetite loss (60%); and minimal for offer comparable overall survival and quality-adjusted
dyspnea (37%), cough (31%), and fatigue (28%) life expectancy as surgery. They concluded by stating
(Langendijk et al. 2001). The global QOL response the need for prospective studies comparing not only
rate improved overall in 36%. While radiographic survival and cost, but also the QOL characteristics of
tumor response was associated with palliation of these treatment modalities (Louie et al. 2010).
symptoms, no significant overall association between A comparative study of radiotherapy versus che-
such response and either palliation of symptoms or motherapy and its impact on QOL was reported by
QOL was demonstrated. kaasa et al. (1989). They reported a QOL study
Similar QOL outcomes were found in an earlier comparing radiation (i.e. 42 Gy in 15 fractions)
study of palliative radiation for inoperable lung can- versus chemotherapy (combination of cisplatin and
cer patients by Langendijk et al. (2000a). After etoposide) in 102 patients with inoperable NSCLC.
curative radiotherapy (i.e. 70 Gy in 35 fractions) in 46 Psychosocial well-being, disease-related symptoms,
patients with medically inoperable Stage I NSCLC, physical function and everyday activity were assessed
Langendijk et al. (2002) found a significant gradual with a validated patient-reported self-administered
increase in dyspnea, fatigue, appetite loss ,and a questionnaire. They found significant differences in
gradual decline for role functioning. As other symp- psycho-social well-being and global QOL that
toms and functioning scales of the EORTC QLQ-C30 favored radiotherapy with no significant differences in
and QLQ-LC30 did not significantly change, they physical functioning or daily activities. The impact of
suggested the observed QOL outcomes may have the addition of chemotherapy to radiation therapy
been a function of pre-existing, slowly progressive versus aggressive radiotherapy alone was recently
chronic obstructive pulmonary disease and radiation- assessed in a study of 75 lung cancer patients
induced pulmonary changes. Balancing the QOL (Pijls-Johannesma et al. 2009). They evaluated the
outcomes with the low incidence of regional recur- evolution of QOL changes using the EORTC QLQ-30
rence and a median survival of 19 months, they and QLQ-LC13. While overall QOL scores initially
concluded that local curative radiation treatment of decreased after the end of RT, the scores returned to
the primary tumor without elective nodal irradiation baseline within 3 months (Pijls-Johannesma et al.
may be the most appropriate treatment for medically 2009). Significant predictors of QOL were maximal
inoperable patients with small, peripherally located esophageal toxicity ([ or = grade 2), Stage IIIA/B,
tumors. More recently, van der Voort et al. (2010) gender and fatigue.
reported QOL outcomes on 39 patients with medi- RTOG 9801 was a study designed to test whether a
cally inoperable Stage I peripherally located NSCLC radioprotector (i.e. amifostine) would reduce the rate
tumors treated with stereotactic radiotherapy (i.e. of chemoradiation-associated esophagitis, and thus
60 Gy in 3 fractions). They evaluated the dynamics of improve QOL in patients with locally-advanced
670 M. Salim Siddiqui et al.

NSCLC (Movsas et al. 2005). In this study, 243 physician-reported assessments underestimated
patients with Stage II or IIIA/B NSCLC received patient distress, particularly in the case of pain (Basch
treatment consisting of induction paclitaxel, followed et al. 2006, 2009; Jensen et al. 2006). This ‘‘discon-
by concurrent weekly paclitaxel and carboplatin with nect’’ may provide an opportunity for a new approach
hyperfractionated RT (69.6 Gy in fractions of 1.2 Gy in adverse symptom reporting. Basch et al. (2009)
given twice per day), starting day 43. Patients were studied 163 patients with lung cancer receiving che-
randomized at registration to either receive amifostine motherapy and used a time-dependent Cox regression
(500 mg IV) four times per week or not receive it model to compare PROs and physician-reported out-
during chemoradiotherapy. Toxicity was assessed comes relating to sentinel clinical events. They found
using NCI-Common toxicity criteria (NCI-CTC), that physician-reported CTCAE assessments were
physician dysphagia logs (PDLs), and daily patient better predictors of unfavorable clinical events;
swallowing diaries. The EORTC-QLQ C30 and QLQ- however, PROs were better predictors of daily health
LC13 were utilized to collect prospective QOL status. Effectively, they concluded that the PRO and
information. Each arm had comparable baseline physician-reported outcomes were complementary
demographics. The median survival rates were com- clinically meaningful data sets. The implication of
parable between both treatment arms (17.3 months this result would be that both data sets should be
with amifostine vs. 17.9 months without amifostine, collected in clinical trials, or that these approaches
p = 0.87). Interestingly, Movsas et al. (2009) recently should be combined.
reported that patient-reported baseline QOL in this
study was an independent prognostic factor for
overall survival, replacing traditional prognostic fac- 2 Conclusions and Future
tors, such as KPS and stage. While others [e.g. Directions
(Langendijk et al. 2000b)] have demonstrated this
prognostic value in patients receiving palliative Lung cancer remains a leading cause of cancer death
radiotherapy, this study (Movsas et al. 2009) was the for both men and women. Even with advances in
first to report this prognostic significance in patients diagnostics and treatments, clinical trials in lung
treated with chemoradiotherapy. Interestingly, RTOG cancer should not only evaluate traditional outcome
9801 demonstrated no significant difference Grade C3 parameters, such as survival and local control, but
esophagitis rates (i.e. 30% with amifostine vs. 34% also investigate QOL measures. As the number of
without amifostine, p = 0.9). However, based on QOL studies in lung cancer continue to grow, certain
patient swallowing diaries, amifostine significantly challenges will need to be addressed. Particularly in
lowered swallowing dysfunction during chemoradia- an era of increasing electronic medical records, new
tion (Z-test, p = 0.03), especially among females electronic, privacy-secure, and internet-based pro-
(p = 0.006) and patients [65 years (p = 0.003). In grams should allow more efficient and real-time QOL
addition, based on QOL forms, patients receiving data collection and tracking. Hopefully, such pro-
amifostine demonstrated less deterioration in patient- grams will improve compliance and minimize data
reported pain at 6 weeks of follow-up (versus pre- management and resource burdens. Secondly, given
treatment) (p = 0.003). Thus, while there was no the clinical relevance of PROs, the NIH has devel-
significant difference between NCI-CTC physician- oped an initiative, Patient-Reported Outcomes Mea-
reported esohpagitis, PRO suggested that amifostine surement Information System (PROMIS) to develop
had some benefit. This result highlights ‘‘disconnect’’ brief QOL instruments to capture patient-reported
between the PRO (e.g. the swallowing diaries/QOL symptoms and PROs in clinical practice (Garcia et al.
forms in RTOG 9801) and the physician-reported 2007). Such initiatives should afford a dynamic series
information (e.g. CTCAE toxicity measures) (Sarna of instruments to measure PROs. Thirdly, while much
et al. 2008). This not only suggests that current clin- work has been done testing the validity and reliability
ical toxicity endpoints may not be sensitive enough, of current QOL instruments, evolution in treatment
but also may primarily reflect the physician rather techniques will require continued refinement for more
than the patient perspective. Such ‘‘disconnect’’ has focused and validated QOL instruments. Another
been observed in other studies, where again challenge centers on understanding the clinical
Quality of Life Outcomes in Radiotherapy of Lung Cancer 671

relevance of QOL outcomes as treatments continue to Burris HA 3rd, Moore MJ et al (1997) Improvements in
evolve. Future studies are beginning to elucidate the survival and clinical benefit with gemcitabine as first-line
therapy for patients with advanced pancreas cancer: a
biologic underpinnings supporting the relationship randomized trial. J Clin Oncol 15(6):2403–2413
between QOL and survival (Sprangers et al. 2009). Cella DF, Tulsky DS (1990) Measuring quality of life today:
Regardless of these challenges, the most compelling methodological aspects. Oncology (Williston Park)
reason to assess QOL and the impetus to address these 4(5):29–38 discussion 69
Cella DF, Tulsky DS et al (1993) The functional assessment of
challenges will still center on patient themselves, and cancer therapy scale: Development and validation of the
their clear desire to discuss QOL issues (Detmar et al. general measure. J Clin Oncol 11(3):570–579
2002; Velikova et al. 2004). Ultimately, QOL will Cella DF, Bonomi AE et al (1995) Reliability and validity of
become a routine clinical tool in the art of caring for the functional assessment of cancer therapy-lung (FACT-L)
quality of life instrument. Lung Cancer 12(3):199–220
our patients. Cella D, Eton DT et al (2002) What is a clinically meaningful
change on the functional assessment of cancer therapy-lung
(FACT-L) questionnaire? Results from Eastern Cooperative
References Oncology Group (ECOG) Study 5592. J Clin Epidemiol
55(3):285–295
Coates A, Porzsolt F et al (1997) Quality of life in oncology
Aaronson N, Fayers P (eds) (2002) Quality of life. Oxford practice: Prognostic value of EORTC QLQ-C30 scores in
textbook of oncology. Oxford University Press, Oxford patients with advanced malignancy. Eur J Cancer 33(7):
Aaronson NK, Ahmedzai S et al (1993) The European 1025–1030
organization for research and treatment of cancer QLQ- Cohen J (1988) Statistical power analysis for the behavioral
C30: a quality-of-life instrument for use in international sciences. Erlbaum Associates, Hillsdale, N. J, L
clinical trials in oncology. J Natl Cancer Inst 85(5): Curran D, Molenberghs G et al (1998) Incomplete quality of
365–376 life data in randomized trials: Missing forms. Stat Med
Basch E, Iasonos A et al (2006) Patient versus clinician 17(5–7):697–709
symptom reporting using the national cancer institute Dancey J, Zee B et al (1997) Quality of life scores: an independent
common terminology criteria for adverse events: results of prognostic variable in a general population of cancer patients
a questionnaire-based study. Lancet Oncol 7(11):903–909 receiving chemotherapy. The national cancer institute of
Basch E, Jia X et al (2009) Adverse symptom event reporting canada clinical trials group. Qual Life Res 6(2):151–158
by patients vs clinicians: relationships with clinical out- de Graeff A, de Leeuw JR et al (2001) Sociodemographic
comes. J Natl Cancer Inst 101(23):1624–1632 factors and quality of life as prognostic indicators in head
Beller E, Tattersall M et al (1997) Improved quality of life with and neck cancer. Eur J Cancer 37(3):332–339
megestrol acetate in patients with endocrine-insensitive Detmar SB, Muller MJ et al (2002) Health-related quality-of-
advanced cancer: a randomised placebo-controlled trial. life assessments and patient–physician communication: a
Australasian megestrol acetate cooperative study group. randomized controlled trial. Jama 288(23):3027–3034
Ann Oncol 8(3):277–283 Efficace F, Bottomley A (2005) Toward a clearer understanding
Bergman B, Aaronson NK et al (1994) The EORTC QLQ- of the prognostic value of health-related quality-of-life
LC13: a modular supplement to the EORTC core quality of parameters in breast cancer. J Clin Oncol 23(6):1335–1336
life questionnaire (QLQ-C30) for use in lung cancer clinical author reply 1336
trials. EORTC study group on quality of life. Eur J Cancer Erridge SC, Gaze MN et al (2005) Symptom control and quality
30A(5):635–642 of life in people with lung cancer: a randomised trial of two
Bergner M, Bobbitt RA et al (1976) The sickness impact palliative radiotherapy fractionation schedules. Clin Oncol
profile: Validation of a health status measure. Med Care (R Coll Radiol) 17(1):61–67
14(1):57–67 Fairclough DL, Peterson HF et al (1998) Comparison of several
Bergner M, Bobbitt RA et al (1981) The sickness impact model-based methods for analysing incomplete quality of
profile: Development and final revision of a health status life data in cancer clinical trials. Stat Med 17(5–7):781–796
measure. Med Care 19(8):787–805 Fang FM, Liu YT et al (2004) Quality of life as a survival
Bezjak A, Dixon P et al (2002) Randomized phase III trial of predictor for patients with advanced head and neck carci-
single versus fractionated thoracic radiation in the palliation noma treated with radiotherapy. Cancer 100(2):425–432
of patients with lung cancer (NCIC CTG SC.15). Int J Fayers PM, Curran D et al (1998) Incomplete quality of life
Radiat Oncol Biol Phys 54(3):719–728 data in randomized trials: Missing items. Stat Med 17(5–7):
Blazeby JM, Brookes ST et al (2001) The prognostic value of 679–696
quality-of-life scores during treatment for oesophageal Food and Drug Administration (2009) Guidance for industry:
cancer. Gut 49(2):227–230 Patient-reported outcome measures: use in medical product
Bleehen NM, Girling DJ et al (1991) Inoperable non-small-cell development to support labeling claims. http://www.
lung cancer (NSCLC): a medical research council random- fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
ised trial of palliative radiotherapy with two fractions or ten Information/Guidances/UCM193282.pdf
fractions. Report to the medical research council by its lung Ganz PA, Figlin RA et al (1989) Supportive care versus
cancer working party. Br J Cancer 63(2):265–270 supportive care and combination chemotherapy in
672 M. Salim Siddiqui et al.

metastatic non-small cell lung cancer. Does chemotherapy Leplege A, Hunt S (1997) The problem of quality of life in
make a difference?. Cancer 63(7):1271–1278 medicine. Jama 278(1):47–50
Garcia SF, Cella D et al (2007) Standardizing patient-reported Little RJA, Rubin DB (2002) Statistical analysis with missing
outcomes assessment in cancer clinical trials: a patient- data. Hoboken, N.J, Wiley
reported outcomes measurement information system initia- Louie AV, Rodrigues G et al (2010) Stereotactic body
tive. J Clin Oncol 25(32):5106–5112 radiotherapy versus surgery for medically operable Stage I
Gelber RD, Gelber S (1995) Quality-of-life assessment in non-small-cell lung cancer: a markov model-based deci-
clinical trials. Cancer Treat Res 75:225–246 sion analysis. Int J Radiat Oncol Biol Phys. doi:10.1016/
Gotay CC (2004) Assessing cancer-related quality of life across j.ijrobp.2010.06.040
a spectrum of applications. J Natl Cancer Inst Monogr Macbeth FR, Bolger JJ et al (1996) Randomized trial of
2004(33):126–133 palliative two-fraction versus more intensive 13 fraction
Guyatt GH, Osoba D et al (2002) Methods to explain the radiotherapy for patients with inoperable non-small cell
clinical significance of health status measures. Mayo Clin lung cancer and good performance status. Medical research
Proc 77(4):371–383 council lung cancer working party. Clin Oncol (R Coll
Hahn EA, Webster KA et al (1998) Missing data in quality of Radiol) 8(3):167–175
life research in Eastern Cooperative Oncology Group McHorney CA, Ware JE Jr et al (1993) The MOS 36 Item
(ECOG) clinical trials: Problems and solutions. Stat Med short-form health survey (SF-36): II. Psychometric and
17(5–7):547–559 clinical tests of validity in measuring physical and mental
Hollen PJ, Gralla RJ et al (1993) Quality of life assessment in health constructs. Med Care 31(3):247–263
individuals with lung cancer: Testing the lung cancer McHorney CA, Ware JE Jr et al (1994) The MOS 36 item
symptom scale (LCSS). Eur J Cancer 29A(Suppl 1):S51–S58 short-form health survey (SF-36): III. Tests of data quality,
Hollen PJ, Gralla RJ et al (1994a) Quality of life during clinical scaling assumptions, and reliability across diverse patient
trials: Conceptual model for the Lung Cancer Symptom groups. Med Care 32(1):40–66
Scale (LCSS). Support Care Cancer 2(4):213–222 Miller GA (1956) The magical number seven plus or minus
Hollen PJ, Gralla RJ et al (1994b) Measurement of quality of two: Some limits on our capacity for processing informa-
life in patients with lung cancer in multicenter trials of new tion. Psychol Rev 63(2):81–97
therapies. Psychometric assessment of the lung cancer Moinpour CM, Feigl P et al (1989) Quality of life end points in
symptom scale. Cancer 73(8):2087–2098 cancer clinical trials: Review and recommendations. J Natl
Husted JA, Cook RJ et al (2000) Methods for assessing Cancer Inst 81(7):485–495
responsiveness: a critical review and recommendations. Moinpour CM, Sawyers Triplett J et al (2000) Challenges
J Clin Epidemiol 53(5):459–468 posed by non-random missing quality of life data in an
Jensen K, Bonde Jensen A et al (2006) The relationship advanced-stage colorectal cancer clinical trial. Psychoon-
between observer-based toxicity scoring and patient cology 9(4):340–354
assessed symptom severity after treatment for head and Montazeri A, Milroy R et al (2001) Quality of life in lung
neck cancer. A correlative cross sectional study of the cancer patients: as an important prognostic factor. Lung
DAHANCA toxicity scoring system and the EORTC quality Cancer 31(2–3):233–240
of life questionnaires. Radiother Oncol 78(3):298–305 Movsas B (2003) Quality of life in oncology trials: a clinical
Johnson JR, Temple R (1985) Food and drug administration guide. Semin Radiat Oncol 13(3):235–247
requirements for approval of new anticancer drugs. Cancer Movsas B, Scott C et al (2005) Randomized trial of amifostine
Treat Rep 69(10):1155–1159 in locally advanced non-small-cell lung cancer patients
Kaasa S, Mastekaasa A et al (1989) Prognostic factors for patients receiving chemotherapy and hyperfractionated radiation:
with inoperable non-small cell lung cancer, limited disease. radiation therapy oncology group trial 98–01. J Clin Oncol
The importance of patients’ subjective experience of disease 23(10):2145–2154
and psychosocial well-being. Radiother Oncol 15(3):235–242 Movsas B, Moughan J et al (2009) Quality of life supersedes
Langendijk H, Aaronson NK et al (2000a) The prognostic the classic prognosticators for long-term survival in locally
impact of quality of life assessed with the EORTC QLQ- advanced non-small-cell lung cancer: an analysis of RTOG
C30 in inoperable non-small cell lung carcinoma treated 9801. J Clin Oncol 27(34):5816–5822
with radiotherapy. Radiother Oncol 55(1):19–25 NCI, C. T. C. G. P (1988) Cancer therapy evaluation program:
Langendijk JA, ten Velde GP et al (2000b) Quality of life after Guidelines. N. C. I. Division of Cancer Treatment,
palliative radiotherapy in non-small cell lung cancer: a Bethesda, MD
prospective study. Int J Radiat Oncol Biol Phys 47(1):149–155 Nicklasson M, Bergman B (2007) Validity, reliability and
Langendijk JA, Aaronson NK et al (2001) Prospective study on clinical relevance of EORTC QLQ-C30 and LC13 in
quality of life before and after radical radiotherapy in non- patients with chest malignancies in a palliative setting.
small-cell lung cancer. J Clin Oncol 19(8):2123–2133 Qual Life Res 16(6):1019–1028
Langendijk JA, Aaronson NK et al (2002) Quality of life after Norman GR, Sloan JA et al (2003) Interpretation of changes
curative radiotherapy in Stage I non-small-cell lung cancer. in health-related quality of life: the remarkable univer-
Int J Radiat Oncol Biol Phys 53(4):847–853 sality of half a standard deviation. Med Care 41(5):582–
Leitgeb C, Pecherstorfer M et al (1994) Quality of life in 592
chronic anemia of cancer during treatment with recombi- Nunnaly J, Bernstein I (1994) Psychometric therapy. McGraw-
nant human erythropoietin. Cancer 73(10):2535–2542 Hill, Mew York
Quality of Life Outcomes in Radiotherapy of Lung Cancer 673

Osoba D (1996) Rationale for the timing of health-related Temel JS, Greer JA et al (2010) Early palliative care for
quality-of-life (HQL) assessments in oncological palliative patients with metastatic non-small-cell lung cancer. N Engl
therapy. Cancer Treat Rev 22(Suppl A):69–73 J Med 363(8):733–742
Osoba D, Rodrigues G et al (1998) Interpreting the significance Testa MA, Simonson DC (1996) Assesment of quality-of-life
of changes in health-related quality-of-life scores. J Clin outcomes. N Engl J Med 334(13):835–840
Oncol 16(1):139–144 Troxel AB, Fairclough DL et al (1998) Statistical analysis of
Pijls-Johannesma M, Houben R et al (2009) High-dose quality of life with missing data in cancer clinical trials. Stat
radiotherapy or concurrent chemo-radiation in lung cancer Med 17(5–7):653–666
patients only induces a temporary, reversible decline in van der Voort van Zyp NC, Prevost JB et al (2010) Quality of
QoL. Radiother Oncol 91(3):443–448 life after stereotactic radiotherapy for stage I non-small-cell
Qian W, Parmar MK et al (2000) Analysis of messy longitu- lung cancer. Int J Radiat Oncol Biol Phys 77(1):31–37
dinal data from a randomized clinical trial. MRC lung Velikova G, Booth L et al (2004) Measuring quality of life in
cancer working party. Stat Med 19(19):2657–2674 routine oncology practice improves communication and
Ribaudo HJ, Thompson SG (2002) The analysis of repeated patient well-being: a randomized controlled trial. J Clin
multivariate binary quality of life data: a hierarchical model Oncol 22(4):714–724
approach. Stat Methods Med Res 11(1):69–83 Ware JE Jr, Sherbourne CD (1992) The MOS 36 item short-
Rubin DB (1997) Estimating causal effects from large data sets form health survey (SF-36). I. Conceptual framework and
using propensity scores. Ann Intern Med 127(8 Pt 2):757– item selection. Med Care 30(6):473–483
763 Ware JE, Gandek B et al (1996) Evaluating instruments used
Sadura A, Pater J et al (1992) Quality-of-life assessment: cross-nationally: Methods from the IQoLA project. Quality
Patient compliance with questionnaire completion. J Natl of life and pharmacoeconimics in clinical trials. Raven
Cancer Inst 84(13):1023–1026 Press, B. Spilker. New York
Sales AE, Plomondon ME et al (2004) Assessing response bias Watkins-Bruner D, Scott C et al (1995) RTOG’s first quality of
from missing quality of life data: the Heckman method. life study–RTOG 90–20: a phase II trial of external beam
Health Qual Life Outcomes 2:49 radiation with etanidazole for locally advanced prostate
Sarna L, Swann S et al (2008) Clinically meaningful differences cancer. Int J Radiat Oncol Biol Phys 33(4):901–906
in patient-reported outcomes with amifostine in combina- WHO (1996) Quality of life assessment. The WHOQOL group,
tion with chemoradiation for locally advanced non-small- 1994. What quality of life? The WHOQOL group. World
cell lung cancer: an analysis of RTOG 9801. Int J Radiat Health Forum. WHO, Geneva 1996
Oncol Biol Phys 72(5):1378–1384 Wyrwich KW (2004) Minimal important difference thresholds
Slevin ML, Plant H et al (1988) Who should measure quality and the standard error of measurement: is there a connec-
of life, the doctor or the patient?. Br J Cancer 57(1): tion?. J Biopharm Stat 14(1):97–110
109–112 Wyrwich KW, Nienaber NA et al (1999a) Linking clinical
Sprangers MA, Sloan JA et al (2009) The establishment of the relevance and statistical significance in evaluating intra-
GENEQOL consortium to investigate the genetic disposi- individual changes in health-related quality of life. Med
tion of patient-reported quality-of-life outcomes. Twin Res Care 37(5):469–478
Hum Genet 12(3):301–311 Wyrwich KW, Tierney WM et al (1999b) Further evidence
Stewart AL, Ware JE (1992) Measuring functioning and well- supporting an SEM-based criterion for identifying mean-
being: the medical outcomes study approach. Duke Uni- ingful intra-individual changes in health-related quality of
versity Press, Durham life. J Clin Epidemiol 52(9):861–873
Strain JJ (1990) The evolution of quality of life evaluations in Wyrwich KW, Tierney WM et al (2002) Using the standard error
cancer therapy. Oncology (Williston Park) 4(5):22–26 of measurement to identify important changes on the Asthma
discussion 27 quality of life questionnaire. Qual Life Res 11(1):1–7
Sundstrom S, Bremnes R et al (2004) Hypofractionated Wyrwich KW, Bullinger M et al (2005) Estimating clinically
palliative radiotherapy (17 Gy per two fractions) in significant differences in quality of life outcomes. Qual Life
advanced non-small-cell lung carcinoma is comparable to Res 14(2):285–295
standard fractionation for symptom control and survival: a Zee BC (1998) Growth curve model analysis for quality of life
national phase III trial. J Clin Oncol 22(5):801–810 data. Stat Med 17(5–7):757–766
Prognostic Factors in Lung Cancer
Frank B. Zimmermann

Contents Abstract
Lung cancer is a heterogeneous clinical entity of
1 Introduction.............................................................. 676 non-small-cell and small-cell lung cancer types as
2 Non-Small-Cell Lung Cancer................................. 676 well as mixed types, with even different clinical
2.1 Tumor-Related Factors .............................................. 676 behaviors and prognoses within particular patho-
2.2 Patient-Related Factors.............................................. 679 logical subgroups. Current guidelines for treatment
2.3 Treatment-Related Factors ........................................ 681
decisions in NSCLC and SCLC are based on tumor
3 Small-Cell Lung Cancer ......................................... 682 extension and certain additional clinico-histopha-
3.1 Tumor-Related Factors .............................................. 682 tological parameters only. Besides this, numerous
4 Patient-Related Factors........................................... 683 clinical laboratory tests and investigations of the
4.1 Performance Status (Karnofsky Index cellular, molecular and genetic biology of lung
and Weight-Loss) ...................................................... 683
cancer and the environment have been introduced
4.2 Gender and Age......................................................... 684
4.3 Laboratory, Hematological and Immunological into routine in pathology, clinical chemistry and
Factors........................................................................ 684 even nuclear medicine, describing the tumor better
5 Treatment-Related Factors..................................... 684 than ever before. This knowledge might support
the treatment decision, research design and anal-
References.......................................................................... 685
ysis, and may support the development of complex
risk models for prediction of lung cancer mortality,
and for the selection of an appropriate treatment
for each individual case. Unfortunately, countless
articles describing more than 150 different prog-
nostic factors but with enormous heterogeneity by
interstudy variations, patient selection bias, low
number of patients in most trials, retrospective
nature of the trials, and poor statistical power may
cause confusion. The main purpose of this article
with special focus on cancer related, patient
related, and environmental aspects is to allow an
impression on most important and significant
prognostic factors, and to offer a basis for
treatment decision in clinical practice concerning
patients with small-cell and non-small-cell lung
F. B. Zimmermann (&) cancer. Most of those factors should not be taken
University Hospital Basel, Basel, Switzerland into account for treatment selection, but to
e-mail: FZimmermann@uhbs.ch

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 675
DOI: 10.1007/174_2011_308,  Springer-Verlag Berlin Heidelberg 2011
676 F. B. Zimmermann

carefully use them for stratification in prospective treatment decision, research design and analysis, and
clinical trials. It should always be kept in mind, health policy development (Brundage et al. 2001;
that the development of new substances in medical MacKillop 2001; Goldstraw et al. 2011). There is
oncology, and the improvements in both surgical increasing interest to develop complex risk models
and radiation oncology increase the therapeutic for prediction of lung cancer mortality, and for
window, and will change the value of some of the the selection of an appropriate treatment for each
mentioned prognostic factors in near future. individual case.
Countless articles have been published, including a
lot of reviews, describing more than 150 different
prognostic factors, and with increasing focus on
1 Introduction molecular and biological markers. Nevertheless, it
must be considered that the literature is markedly
Lung cancer is a heterogeneous clinical entity, con- heterogeneous with inter-study variations, patient
taining the broad spectrum of non-small cell lung selection bias, low number of patients in most trials,
cancer including not otherwise specified cancer, and retrospective nature of the trials, and poor statistical
the group of small-cell-cancer, as well as mixed types. power (Brundage et al. 2002; Ou et al. 2007;
All groups share molecular and cellular origins, but Mandrekar et al. 2006; Goldstraw et al. 2011).
have distinct clinical behaviors and prognoses, even Therefore, the main purpose of the following
within their particular pathological subgroups. overview is to allow an impression on most important
The afflicted patients present a diverse constella- and significant prognostic factors, and to offer a basis
tion of clinical symptoms and biochemical values, in for treatment decision in clinical practice concerning
part caused by different manifestations of the primary patients with small-cell and non-small-cell lung
tumor, distinct distribution of involved metastatic cancer.
sites, and a varied extent of paraneoplastic syn- In principle, prognostic factors can be divided into
dromes, and even comorbidities. Inspite of the three subgroups (Goldstraw et al. 2011):
remarkable predictability of population survival • Cancer related as tumor type, size, site, and dif-
outcomes this knowledge is of limited value for ferentiation, and extent of disease.
treatment decisions in a single patient, due to the • Patient-related as comorbidities, gender, perfor-
marked heterogeneity of the clinical course in the mance status, individual habits as smoking.
individual patient (Brundage et al. 2002). Current • Environmental factors as treatment options, social
guidelines for treatment decisions in NSCLC and support, available treatment.
SCLC are based on tumor extension (mainly TNM These factors can be used individually, and may
staging system) and certain additional clinico- create a formula of a composite prognostic factor,
histopathological parameters only. influenzing the treatment decision among other
Besides this, there is a need for an explanation and things.
a new classification of the heterogeneous nature of the
disease and, thereby, select the best individual treat-
ment by the prognosis for each patient. In the previ- 2 Non-Small-Cell Lung Cancer
ous years, numerous clinical laboratory tests, and,
most recently, investigations of the cellular, molecu- 2.1 Tumor-Related Factors
lar and genetic biology of lung cancer and the
environment have been introduced into routine in 2.1.1 Tumor Stage
pathology, clinical chemistry, and even nuclear The definition of major clinical subgroups on the
medicine, describing the tumor better than ever before basis of tumor stage (TNM staging system) has been
(Buccheri and Ferrigno 2004; Feld et al. 1994; consistently shown to be the strongest determinate of
Brundage et al. 2002; Goldstraw et al. 2011; Filosso the outcome of NSCLC patients overall. Within
et al. 2011; Paesmans et al. 2010). This knowledge this system each single parameter describing the
may play an essential role in explaining the different anatomic burden of disease (T = local extent of
outcomes of the patients, and might support the tumor; N = site of nodal metastases; M = number
Prognostic Factors in Lung Cancer 677

and location of distant metastases) reflects prognosis cell type only in stage III A disease, whereas the Lung
(Buccheri and Ferrigno 2004; Ou et al. 2007). Revi- Cancer Study Group reported superior postoperative
sions to the TNM-system were made in 1997 and in outcome for patients with squamous cell carcinoma in
2010, to provide greater specificity for patient sub- all but stage III disease (Mountain et al. 1987). From the
groups, recognizing the prognostic relevant difference preponderant studies, it can be concluded that squa-
even between subgroups of T1 (pT1a: \2 cm; pT1b: mous cell type has a small but independent positive
[2–3 cm) and T2 (pT2a: [3–5 cm; pT2b:[5–7 cm), impact on survival prognosis, and adenosquamous
the importance of tumor-related factors (e.g., cN2 or carcinomas have a comparably poor prognosis (Filosso
cT4 disease) that estimate the likelihood of definitive et al. 2011). In summary, the histological subtype of
resectability within stage III, and of the presence of NSCLC has lower prognostic information in NSCLC
intrapulmonary satellite tumor metastases in a dif- than stage, age, performance status and gender, and this
ferent ipsilateral lobe from that of the primary (T4) might additionally be influenced by a different grading
(Goldstraw et al. 2011; Sobin et al. 2010). Besides and rate of apoptosis and mitosis as risk factors for
T-category, it is known that tumor size is the powerful distant metastasis at least (Komaki et al. 1996; Ou et al.
predictor of survival in patients who have disease that 2007).
is amenable to resection but who are inoperable due to
medical reasons and will undergo definite radiother- 2.1.3 Biological and Genetic Factors
apy (Wigren et al. 1997; Ou et al. 2007; Sobin et al. Biological and genetic tumor factors have been
2010; Olson et al. 2011). evaluated mostly in resected specimen, therefore data
In summary, the anatomical extent of the tumor, on patients with advanced disease are rare. Never-
defined by the TNM-system, is one of the most theless, some of those factors have been shown to
important prognostic factors, the most accurate and have independent prognostic significance. They are
reliable way to estimate the patients perspective (Li rarely assessed in clinical routine practice: histologic
et al. 2011a). Therefore, accurate clinical assessment features, markers of tumor proliferation, markers of
of the TNM stage is of utmost importance (Birim cellular adhesion, and other molecular biological
et al. 2006). However, it cannot predict precisely the markers.
5-year survival rate even in fictitious homogeneous The latter group includes regulators of cellular
early stage tumors (mean value 67%) (Mountain growth (e.g., ras oncogene or protein, retinoblastoma,
1997; Kwiatkowski et al. 1998). This indicates that epidermal growth factor receptor EGFR and EGFR
the TNM-system based on clinical, radiological, copy numbers well as EGFR gene mutations, erb-b2,
invasive and even histopathological investigations is motility-related protein-1, and hepatocyte growth fac-
far from sufficient. It might be explained by problems tor), regulators of the cell cycle and apoptosis (p53, bcl-
in staging procedures, but will also be influenced by 2, KRAS, p27), the nucleotide excision repair pathway
other prognostic factors, tumor or patient related, (excision repair cross-complementation group 1
having been studied in addition to anatomical extent ERCC1, ribonucleotide reductase messenger 1 RRM1,
by the IASLC staging project (Sculier et al. 2008). breast cancer gene 1 BRCA1), and regulators of the
metastatic cascade (e.g., tissue polypeptide antigen
2.1.2 Tumor Histology [TPA], cyclin D-1, and cathepsin).
The prognostic significance of histopathological cell Of those factors, especially EGFR mutations play
type of NSCLC (e.g., large-cell, adenocarcinoma, an important prognostic and predictive role, with
squamous cell, undifferentiated cancer) has been superior treatment outcomes in mutation-positive
studied extensively. Some authors did not find a prog- subgroups receiving tyrosine-kinase-inhibitors of
nostic implication of cell subtype, but the majority of EGFR (EGFR-TKI). However, response to EGFR-
retrospective trials have shown an independent supe- TKI is not exclusive to patients with EGFR-mutation-
rior outcome in squamous cell carcinoma (Komaki positive tumors, and the impact on overall survival is
et al. 1996; Birim et al. 2006), with an advantage of not completely clear. Further prospective clinical
about 10% in 5-year survival rate. Unfortunately, the trials focus on this topic (Rossi et al. 2009).
results are conflicting, with Sculier et al. (2008) p21 status, status of the serum assay for detection
reporting a significant prognostic value of histology of the cytokeratin 19 fragment, status of the
678 F. B. Zimmermann

argyrophilic nucleolar organizer region, and p185 importance of subgroups of VEGF has been validated
status have been significantly associated with prog- by a recent systematic review with metaanalysis in
nosis in about 80% of the studies, whereas Ki-67 2009 (Zhan et al. 2009). A higher level of VEGF in
status, vascular endothelial growth factor status, and blood and tumor tissue is strongly correlated with a
vessel invasion were positive correlated in only poorer outcome of the patient. Assessment of circu-
50–60%. Data on mutation of the p53 suppressor gene lating levels of VEGF may be valuable future tools
are also conflicting, but several systematic reviews for treatment planning and monitoring treatment
have confirmed its prognostic impact at least in effects and tumor relapse (Bremnes et al. 2006). Also,
adenocarcinoma. Unfortunately, only a few reports both high VEGF-A and VEGF-C protein expression
are based on prospectively designed studies thus have been associated with poor survival in the
decreasing their value (Buccheri and Ferrigno 2004; majority of clinical trials on NSCLC (Reinmuth et al.
Coate et al. 2009). And, so far, none of these factors 2010). With an increasing number of biomarker
can be really used for treatment decision. The ques- studies there may be a chance to better characterize
tion of which molecular markers will prove to be the the tumor, and thereby tailor an individualized perfect
most useful for selecting treatment for individual treatment for each patient. Unfortunately, other types
patients remains unanswered. of growth factors (VEGFR2, VEGFR3) seem not to
Therefore, although well-developed and validated have an impact on prognosis and will not help in
genomic signatures could lead to personalized treat- treatment decision, whilst for further markers as
ment decisions, the practicing physician and the bFGF (basic fibroblast growth factor), hypoxia-
patient are still left in doubt about the reliability and inducible factor (HIF), and metastasis-associated
medical utility of the signatures (Rossi et al. 2009; protein 1 (MTA1) the data are still controversial or
Subramanian and Simon 2010). The importance of too early to be safely interpreted (Bremnes et al.
standardization and prospective validation not only of 2006; Zhan et al. 2009; Bonnesen et al. 2009; Wu
the markers but the techniques used in molecular and et al. 2011; Li et al. 2011b).
biological markers can only be further emphasized, Matrix metalloproteinases 2 and 9 (MMP-2, MMP-
which were immunohistochemistry, gene expression, 9) participate in many deregulated signaling pathways
mutational analysis, and microarray (Goldstraw et al. that are used by the tumor to promote cancer cell
2011). growth, angiogenesis, and apoptosis, especially deg-
radation of proteins in the extracellular matrix and
2.1.4 Angiogenesis, Growth Factors activation of growth factors. In a recent meta-analysis
and Carcinogenic Process on MMP-2 (Qian et al. 2010) and a single-center
It is evident that angiogenesis is a relatively early evaluation on MMP-9 (Martins et al. 2009), the
event during cancer pathogenesis, and it is at least in prognostic value on overall survival either in NSCLC
part responsible to ensure tumor oxygenation, nutri- for MMP-2 or in adenocarcinoma only for MMP-9
tion, and to cause distant metastasis. Neoangiogenesis appeared significant. The integration of MMP-2
as a major basis for tumor growth and metastasis has as a selection marker for a prospective trial to pro-
been evaluated in surgical specimen. Microvessel spectively evaluate its real value was demanded
optical count was done in patients with stage I to IIIA (Qian et al. 2010).
disease, and found to be a powerful independent
prognostic factor (Fontanini et al. 1998). This statis- 2.1.5 Serological Tumor Markers
tical significance was confirmed by a large review of The value of standard tumor markers as predictive
several thousands of patient records by the European parameter has been tested in several clinical trials.
Lung Cancer Working Party in 2002 (Meert et al. Cytokeratin-19-fragments (Cyfra 21-1), tissue poly-
2002). peptide antigen (TPA), cancer antigen 125, and car-
In the previous years, an increasing number of cinoembryonic antigen (CEA) have been evaluated as
angiogenic cytokines have been identified being valid prognostic determants, with cytokeratin frag-
involved in neoangiogenesis of lung cancer, with ments almost certain. Unfortunately, the data are not
vascular endothelial growth factors (VEGF) appar- homogeneous, and the prognostic capability of CEA
ently being the most important one. The prognostic was rather weak (Buccheri and Ferrigno 2004).
Prognostic Factors in Lung Cancer 679

In stage I disease, CEA may be used to indicate scale for geriatrics CIRS-G) (Charlson et al. 1987;
adjuvant chemotherapy, but this has been shown Miller et al. 1992).
reliable only within small retrospective trials (Wang In several former studies the male sex was dis-
et al. 2010; Hsu et al. 2007; Matsuoka et al. 2007). cussed as an adverse prognostic factor. A review in
Cyfra 21-1 has been evaluated in a multivariate 1994 described significant evidence in 7 out of 19
analysis, and was proven with a higher sensitivity to studies with univariate analysis, and in 9 of 23
predetermine the treatment outcome than CEA and studies with multivariate analysis in favor of the
NSE (Picardo et al. 1996; Reinmuth et al. 2002). female sex (Buccheri and Ferrigno 1994). These data
More recent data do not support the value of Cyfra were confirmed by an evaluation in a tumor register
21-1 as a prognostic indicator in stage I (Matsuoka population. Median survival was significantly better
et al. 2007), and, therefore, this marker may be only for women than for men, and together with the
used to control the therapeutic efficacy of chemo- or extent of tumor and weight loss gender was the
radiochemotherapy in more advanced disease. A new strongest independent predictor even in multivariate
marker, the carbohydrate antigen Sialyl Lewisx (SLX) analysis (Palomares et al. 1996). This may be
has been seen significantly linked with overall sur- explained by the higher life expectancy of women
vival in resected stage I NSCLC, but has to be proven regardless of the presence of lung cancer, a lower
prospectively in larger trials (Mizuguchi et al. 2007). incidence of severe comorbidity of heart and lung,
In principle, outside of clinical trials, none of the less tobacco consumption, higher incidence of ade-
tumor markers should be used for treatment decision nocarcinoma, and less frequent pneumonectomies
before initiating therapy, but only for follow-up. (Birim et al. 2006). Furthermore, the hormonal and
metabolic situation in women seems to positively
influence the aggressiveness of lung cancer (Birim
2.2 Patient-Related Factors et al. 2006). This may change with the increasing
incidence of lung cancer in women and the changing
2.2.1 Gender, Age and Ethnicity smoking habits.
Unfortunately, the literature is quite variable in the On the other hand, very recent data from the
conclusions about the prognostic value of gender and Canada Clincal Trials Group indicated only a modest
age, and the strength of the association with survival effect of gender on progression-free survival for
outcomes. Although reports on age in multivariate women under chemotherapy, but no influence on
analysis have been inconsistent, a younger age might overall survival and quality of life, whereas a Japa-
carry a better prognosis (Quejada and Albain 2004). nese analysis pronounced the significant positive
Albain and colleagues identified good performance influence of female gender at least in never-smokers
status, female sex, and age below 70 years as the most (Wheatley-Price et al. 2010; Kawaguchi et al. 2010).
important factors that were predictive of favorable This heterogeneity is in contrast to the more homo-
survival rates overall. Nevertheless, surgical, radia- geneously proven positive impact of female gender in
tion and perioperative intensive care techniques and small-cell lung cancer, and therefore, treatment
knowledge have increased reasonably in the previous decision in NSCLC should not be based on gender.
decades, resulting in lower treatment mortality even The importance of ethnicity and of socioeconomic
in elderly patients. Due to raised comorbidity with status as independent prognosticators has been shown
age, advanced age alone should not be the only reason by a retrospective population-based study from the
to withhold aggressive treatment but the presence of cancer surveillance programs in Southern California.
comorbid conditions (Birim et al. 2006; Asmis et al. Asians had the highest over survival, independent of
2008). Scoring systems for risk assessment before the type of treatment (Ou et al. 2009). A higher
radical surgery and aggressive large-volume radio- socioeconomic status correlated well with a better
therapy would be helpful to better tailor individual outcome, confirmed in a multivariate analysis and
cancer treatment especially for elderly (Charlson thereby not influenced by surgical treatment or mar-
comorbidity index CCI; cumulative illness rating ital status (Ou et al. 2008).
680 F. B. Zimmermann

2.2.2 Performance Status (Karnofsky Index Weight loss within the last 6 months before diag-
and Weight-Loss), Pulmonary Function nosis has an important impact on survival, with total
and Cardiovascular Disease, Smoking proportional weight loss being the most significant one
Habits (Buccheri and Ferrigno 2004; Mandrekar et al. 2006).
Numerous studies investigated patient characteristics In resectable cancer, the prognostic value of pul-
as predictors of survival after surgical resection, monary function has been proven, with the estimation
definite radio- or radiochemotherapy in non-small cell of the absolute and predicted proportional postoper-
lung cancer. Due to the fact that most patients with ative spirometry value being mostly predictive. These
early stage disease are asymptomatic, only few stud- values can be calculated by multiplying the observed
ies have systematically evaluated the prognosis of preoperative value with the percentage of postopera-
patients related to clinical symptoms in stage I cancer. tively remaining lung tissue, or by performing a lung
They have been found to be less powerful predictors perfusion scan (Birim et al. 2006). Cardiovascular
of outcome, particularly in stage I disease, than in the disease has been found in several retrospective trials
advanced disease setting, and, therefore, these factors to be a relevant factor for postoperative morbidity and
are not generally considered to be important for mortality. Therefore, a preoperative evaluation should
clinical decision making. Nevertheless, hemoptysis, be carried out in all patients at risk, using exercise
coughing, and thoracic pain were identified as risk stress tests (Birim et al. 2006).
factors for tumor recurrence and poor survival Actual data from Japan using a database of 4,954
(Harpole et al. 1995). patients showed that never-smoking status was a
In locally advanced and irresectable cancer as well marginal but significant favorable factor regarding
as functional inoperable patients an increasing overall survival, not caused by response on chemo-
amount of research has addressed the use of patient- therapy or influenced by gender or younger age of
reported parameters. The majority of those patients non-smokers. Different profiles of never-smokers with
will show significant symptoms or other general lung cancer will enhance the idea that NSCLC in this
manifestations of illness such as weight loss or poor subgroup may be considered and treated as a dis-
performance status. tinctive disease (Kawaguchi et al. 2010).
Karnofskys index of performance status (KPS) and
the Eastern Cooperative Oncology Group perfor- 2.2.3 Laboratory, Hematological
mance status scale (ECOG PS) have been examined and Immunological Factors
within large trials, and ahead of 50 other factors, KPS Hematologic or biochemical markers might be associ-
and weight loss within the previous 6 months were ated with disease extent, and therefore have been eval-
the most important, besides the extent of disease uated in numerous trials regarding their prognostic
(Stanley 1980; Mandrekar et al. 2006; Li et al. 2011a, value. In a large study of 2,531 patients who were
b). Large clinical studies or reviews confirmed KPS as enrolled in a variety of clinical trials, four prognostic
well as ECOPG PS as one of the two most important factors for patients receiving cisplatin chemotherapy
prognostic factors, with ECOG PS being the more were identified that had significantly distinct survival
reliable and useful (Buccheri and Ferrigno 2004; expectations: performance status, age, and hemoglobin
Kawaguchi et al. 2010). In recent trials, the role of and serum LDH levels. Other studies employing sec-
cancer related symptoms, quality-of-life scores, and/ ondary analysis of clinical trial information or after
or anxiety and depression measures have been retrospective evaluation of patient data outside of clini-
investigated more in depth. Those studies reported the cal protocols have reached similar conclusions (Paes-
importance of quality of life, being a stronger deter- mans et al. 1995; Hespanhol et al. 1995; Takigawa et al.
minant than pure performance status (Langendijk 1996; Mandrekar et al. 2006; Goldstraw et al. 2011).
et al. 2000; Buccheri and Ferrigno 2004). Quality- In general, high white blood cell count, low
of-life scores and anxiety and depression assessments hemoglobin level, and LDH are certainly the strongest
may reflect the extent of disease and also the patients’ prognostic factors, whether considered alone or in
inherent characteristics or degree of emotional sup- combination with weight loss, performance status,
port that may predict better disease outcomes, possi- or tumor stage (Buccheri and Ferrigno 1994;
bly through psychophysiologic mechanisms. Mandrekar et al. 2006; Maione et al. 2009; Goldstraw
Prognostic Factors in Lung Cancer 681

et al. 2011), mainly in advanced stage tumors. Further 5-year survival rate, but not in very early stage NSCLC
independent laboratory tests are calcium and albumin, (p T 1–2 N 0) where it gives the same results as
with decreased values predicting poor prognosis. lobectomy (Graziano 1997; Jazieh et al. 2000; Na-
Thrombocytosis above 400.000/yl, tested in a spe- kamura et al. 2011). It has been clearly documented that
cifically designed study, showed a strong correlation surgical procedure is a significant prognostic factor.
with advanced disease and decreased survival even Of fatal prognostic significance is an incomplete
after adjustment for stage and histological type of resection, either with gross disease remaining or with
tumor, sex and age of the patient (Pedersen and positive microscopic resection margins, even when
Milman 1996). Based on these data, Mandrekar et al. additional postoperative therapy (radiotherapy or che-
(2006) created a prediction equation for the prognosis moradiotherapy) is provided (Ginsberg et al. 1999),
of patients with newly diagnosed stage IV NSCLC, suggesting the poor biological characteristics of the
useful to design trials and compare results of phase-II tumor being both associated with locoregional extension
trials with each other. that causes microscopic residual disease and early sys-
temic spread. Perioperative blood transfusions, required
2.2.4 Standardized Uptake Value mainly in extensive dissections, is postulated to decrease
in FDG-PET overall survival by mediated immunosuppression
Several large retrospective evaluations (Al-Saraf et al. favoring proliferation, distant spread, and migration of
2008; Agarwal et al. 2010) and an updated meta-anal- tumor cells. Published data are incongruous, with
ysis (Paesmans et al. 2010) have shown SUV being a shortening time to recurrence, overall and recurrence-
very interesting factor for predicting patient outcome, free survival by 30% in some publications, and no sig-
with a strong correlation regarding overall survival and nificant influence in others (Quejada and Albain 2004).
cancer-specific survival as well. The results do not In marginal resectable situations, preoperative
allow concluding to an optimal threshold but only that chemotherapy may be considered, with proven pro-
higher values of SUV imply higher hazards. It seems longed survival rates, especially in patients with
that there is a continuous increase in risk with responding tumors, making it a highly relevant
increasing SUV (Paesmans et al. 2010). prognostic factor (Birim et al. 2006).
Primary radiotherapy with curative intent is mainly
recommended for patients who cannot undergo resec-
2.3 Treatment-Related Factors tion in curative intention, although no modern ran-
domized trials have directly compared surgery to
2.3.1 Clinically Resectable Disease radiotherapy in early stage NSCLC (Ginsberg et al.
Several modern studies compared resection with 1999; Zimmermann et al. 2003, 2010). In this situation,
combined conservative modality treatments in stage it is well known that treatment results depend on total
III NSCLC, showing no superiority of surgery in dose and fractionation schedule, with acceleration and
regard to overall survival. hyperfractionation to biological effective doses of more
Nevertheless, when surgery is considered to be the than 70 Gy producing superior outcome in stage III
standard management of patients who are medically fit disease, whereas hypofractionated schedules with ste-
for thoracotomy, to aim at both high local disease reotactic techniques produce high local control and
control and acceptable overall survival rates (Sabiston cancer-specific survival rates comparable to resection
and Spencer 1995), complete resection is essential (Sause 2001; Jeremic et al. 2002; Saudners et al. 1999;
(Birim et al. 2006). Lobectomy or pneumonectomy are Willner et al. 2002; Choi et al. 2001; Zimmermann et al.
standard approaches, with wedge resection being 2003, 2010). In this situation, total dose should be
reserved for patients in poor condition even in stage I chosen high enough (BED [100 Gy), to result in a
NSCLC, due to inferior results possibly caused by close perfect long-lasting local tumor control of 90% or
resection margins or limited lymph node dissection higher (Olson et al. 2011).
(Sabiston and Spencer 1995; Lee et al. 1999; Birim
et al. 2006; Nakamura et al. 2011). Tumor wedge 2.3.2 Locally Advanced Disease
resection, segmental or atypical resection increases the Patients without clinical symptoms or radiological
risk of local recurrence three to fivefold with a reduced signs of systemic manifestations but irresectable
682 F. B. Zimmermann

disease have been shown in a number of clinical trials and the most powerful prognostic factor in most of the
to have higher survival rates when they receive published series using the IASLC definition (Shepherd
induction chemotherapy followed by radiotherapy, or et al. 2007; Micke et al. 2002; Paesmans et al. 2000;
even better concurrent chemoradiotherapy, compared Jorgensen et al. 1996). The median survival is around
to radiotherapy alone (Stewart and Pignon 1995). 15 months in limited in contrast to about 10 months in
The same subgroup of patients has been shown to extensive disease patients (Yip and Harper 2000), and
experience higher survival rates when treated with this has a major implication on treatment decision.
continuous hyperfractionated and accelerated radio- Besides this two class-system a lot of other prog-
therapy compared to conventional fractionation, and nostic factors that describe the extent of tumor and the
when treated with higher doses of conventional radi- number or location of metastatic sites involved have
ation compared to lower doses (Saudners et al. 1999; been evaluated (vena cava syndrome, pleural effusion
Willner et al. 2002; Choi et al. 2001; Emami and or nodal involvement; involvement of different organs
Perez 1993). In this situation, clinical tumor response like liver, brain, or bone) (Albain et al. 1990;
is a highly significant prognostic factor, as well as Würschmidt et al. 1995; Tamura et al. 1998; Bremnes
tumor volme. Doses as high as justifiable and rea- et al. 2003). Mediastinal involvement and the infil-
sonable should be aimed for, using modern radiation tration of several organs might decrease the prognosis
techniques as IMRT and SBRT at least for a radiation of the patient, as has been demonstrated regarding
boost (Lee et al. 2011; Alexander et al. 2011). long-term survival by a Canadian Group (Tai et al.
The role of surgery in relation to induction che- 2003), but data are not consistent. Therefore, these
motherapy and radiotherapy is still investigational, as factors are not generally used as a basis for treatment
is the role of combination chemoradiotherapy in more decision.
symptomatic patients (Ginsberg et al. 1999). Never-
theless, in some situations neoadjuvant chemotherapy 3.1.2 Histological Subtypes
can be considered, with increasing prognosis at least Small-cell lung cancer can carry a mixture of different
for responders (Birim et al. 2006). tumor cells in upto 20% of the cases, large-cell car-
cinoma being the most commonly combined cell type.
2.3.3 Metastatic Disease This causes the pathological committee of the IASLC
For patients without substantial systemic manifesta- to adopt three new subtypes of small-cell lung cancer:
tions of illness and under good condition (Karnofsky small cell, mixed large and small cell, and combined
Index [60), chemotherapy is known to improve small cell carcinomas (Hirsch et al. 1988). Unfortu-
median survival time when compared to the best nately, several following studies could not document
supportive care alone (Stewart and Pignon 1995). a different clinical outcome for these three subgroups,
This has not been documented for patients with poor and the actual WHO classification abandoned the idea
performance status, where best supportive care is of different subgroups (Brambilla et al. 2001).
recommended in general. Nevertheless, the high percentage of patients with
various combinations of small- and non-small cell
lung cancer might explain the divergent response to
3 Small-Cell Lung Cancer chemotherapy, and support the idea of salvage
resection for locally low-responding cancer (Sheperd
3.1 Tumor-Related Factors et al. 1991). No histological factors are predictive of
prognosis in SCLC till now (Fissler-Eckhoff 2010).
3.1.1 Tumor Stage
In contrast to NSCLC, small-cell lung cancer is gen- 3.1.3 Serological Factors (Tumor Markers)
erally classified into a two-stage system—limited and Besides the tumor extent, simple laboratory parame-
extensive disease—with limited disease (tumor con- ters like biochemical tests and serum tumor markers
fined to one hemithorax (Veterans Administration have their predictive values.
Lung Study Group VALG) or without distant metas- Serological factors (tumor markers), produced by
tases (International Association for the Study of Lung tumor cells and released into the bloodstream,
Cancer IASLC), respectively, being tested a definite have been evaluated in a number of different studies.
Prognostic Factors in Lung Cancer 683

Due to their low tumor-specificity only a few of them ratio [5) is correlated with a survival advantage.
have certain prognostic value: neuron specific enolase Unfortunately, in contrast to the prognostic value of
(NSE) and cytokeratin-19-fragments (Cyfra 21-1). p53 antibodies in NSCLC the results from several
NSE has been tested in several large trials, and a actual clinical trials are contradictory, so that the
significant correlation was found between elevated value as prognosticator is not proven certain (Jassem
NSE levels and poor prognosis both in univariate and et al. 2001; Murray et al. 2000).
multivariate analyses, making it one of the most Further genetic and biological abnormalities con-
powerful prognostic factors (Bremnes et al. 2003; nected to the pathogenesis of small-cell lung cancer
Jorgensen et al. 1996). Using NSE together with are under investigation, as thymidylate synthase and
performance status of the patient and tumor extent in epidermal growth factor receptor (EGRF), but none
a simple algorithm produces a clearly defined prog- has already been established as a trustful marker
nostic classification that can be used for treatment predicting the prognosis of a patient with small-cell
decision (Jorgensen et al. 1996). lung cancer. At least, it became obvious that due to
Cyfra 21-1 has been the most commonly studied higher TS-levels in SCLC, a treatment with the
cytokeratin, and besides extensive disease and TS-inhibitor pemetrexed is not reasonable (Rossi
increased levels of LDH and NSE, elevated levels to et al. 2009).
more than 3.6 ng/ml significantly indicated a poor
outcome of the patient (Pujol et al. 2003).
Among the plentiful further tested serological 4 Patient-Related Factors
markers only the serum carcinoembryonic antigen
(CEA) has deserved to be mentioned: in univariate 4.1 Performance Status (Karnofsky
analyses its value has been confirmed, whereas Index and Weight-Loss)
chromogranin A (CgA), pro-gastrin releasing peptide
(ProGRP) and creatinine kinase-BB (CPK-BB) have The performance status describes the patients’ ability
not yet been confirmed valid (Ferrigno et al. 1994; of self-care and to perform normal activity including
Lamy et al. 2000; Sunaga et al. 1999). his participation in social life. There are two different
schedules in use: the Karnofsky Scale of Performance
3.1.4 Biological and Genetic Factors Status (KPS) (11 levels from 100 to 0) and the Eastern
The genetic deletion of a number of chromosomes is Cooperative Oncology Group Scale of Performance
discussed as the major impulse of the development of Status (ECOG PS) (5 levels). The value of both
human lung cancer, stimulating the activation of schedules has been tested, with the ECOG PS being
proto-oncogenes and the loss of tumor suppressor easier to apply and of better discrimination of
genes. In small-cell lung cancer, the activation of patients’ prognosis (Buccheri et al. 1996). In numer-
genes of the myc family (c-myc, L-myc, N-myc) ous and even very large clinical trials the performance
seems to be notable (Rygaard et al. 1993). The status—independent of the schedule used—was con-
expression depends on tissue type, and corresponds to firmed as a significant prognostic co-factor (Paesmans
the maturity and development of different cell lines. 2004; Buccheri and Ferrigno 1994; Osterlind and
The c-myc oncogene may play an important role for Anderson 1986; Spiegelman et al. 1989; Albain et al.
the differentiation of the cell into many cellular pro- 1990; Rawson and Peto 1990).
cesses (proliferation, differentiation, apoptosis). It is Besides tumor extent and performance status
highly amplified in SCLC cell lines in vivo, indicating weight loss has been identified an important prog-
its relation to tumor progression and aggressiveness of nostic factor also in small-cell lung cancer (Stanley
the tumor (Bergh 1990). In clinical studies a high 1980; Tamura et al. 1998; Bremnes et al. 2003).
amplification of c-myc strongly correlates with tumor A more complex determinant predicting the sur-
progression and a poor outcome of the patient (Salgia vival of patients with small-cell lung cancer is quality
and Skarin 1998). of life, a multi-factorial concept considering all
The value of p53 antibody has been evaluated in physical, psychological, social, and functional status
several clinical trials. It seems possible that the of the patient. Quality of life tests have been tested
presence of a high titer of p53 antibody (titer valid in several clinical studies, but are more difficult
684 F. B. Zimmermann

to establish in clinical routine and, therefore, rarely decision (Bremnes et al. 2003; Quoix et al. 2000;
used besides clinical trials (Naughton et al. 2002; Rawson and Peto 1990; Osterlind and Anderson 1986).
Montazeri et al. 2001; Buccheri 1998). In principle, an elevated LDH should be seen as a
risk factor for poor survival and an indicator of a
larger tumor burden, at least demanding for a com-
4.2 Gender and Age plete assessment of tumor stage. Based on the work of
the Subcommittee for the Management of Lung
Gender was documented as a discriminating factor of cancer in UK on almost 4000 patients, performance
SCLC outcome, with the combination of female sex status, disease stage, and AP, Na, aspartate amino-
and younger age (below 60 years) carrying the best transferase and LDH should be measured in all future
prognoses regarding response rates, median survival, trials to assist comparisons between the clinical trials
and 2-year survival rate. This observation was inde- (Rawson and Peto 1990). Together with performance
pendent of any other relevant prognostic variable status and tumor stage, Na, AP, and LDH have been
(Buccheri and Ferrigno 1994; Wolf et al. 1991; combined as the Manchester Prognostic Score, and
Osterlind and Anderson 1986; Spiegelman et al. 1989; later modified by a Japanese group into a new three-
Albain et al. 1990). It was repeatedly confirmed in group-classification (Kawahara et al. 1999). They can
more recent trials, using different types of chemo- differentiate 1-year survival rates of more than 50% in
therapy (Singh et al. 2005; Paesmans et al. 2000), but the best versus 0% in the worst group, or median
should nevertheless not influence treatment decision survival rates of 16.0 versus 6.6 months, respectively.
alone. These classifications can be used to design clinical
trials and to tailor individual treatment as well.

4.3 Laboratory, Hematological


and Immunological Factors 5 Treatment-Related Factors

There is a long list of laboratory tests evaluated as the The response to treatment has been found to be highly
possible prognostic factors in small-cell lung cancer: significantly on survival of patients treated with
lactate dehydrogenase (LDH), hemoglobin serum chemo- and radiochemotherapy. Complete responders
concentration (Hb), albumin, alkaline phosphatase had a better survival than partial responders, who had
(AP), sodium, calcium, creatininemia, bicarbonates, a superior outcome than non-responders (Lebeau et al.
bilirubinemia, erythrocytes, leucocytes, neutrophilia, 1995; Ray et al. 1998; Paesmans et al. 2000).
and thrombocytes. In several randomized trials it has been docu-
Elevated LDH, tested in 10 of 13 multivariate and mented that simultaneous radiochemotherapy, with
three large trials with more than 500 patients each, is the radiotherapy being administered early in the treatment
strongest hematological prognostic factor with high schedule, will improve the outcome of patients in
accuracy predicting poor outcome. It seems to be even good condition compared to chemo- or radiotherapy
more important than tumor markers (NSE), and is alone, and that altered fractionation of irradiation
recommended by different groups as the cheapest and might further enhance the results (Warde and Payne
most valid marker for small-cell lung cancer as a 1992; Murray et al. 1993; Jeremic et al. 1997; Work
stratification criteria for clinical trials (Quoix et al. et al. 1997; Lebeau et al. 1999; Turrisi et al. 1999;
2000; Rawson and Peto 1990; Osterlind and Anderson Takada et al. 2002). In retrospective trials the
1986). Of all the other factors mentioned before, the importance of total radiation dose has been pro-
results are more or less inhomogeneous: low serum nounced (Tai et al. 2003), but a randomized trial
albumin concentration, normal sodium (Na) and uric comparing higher dose with hyperfractionated sche-
acid levels, decreased plasmatic level of hemoglobin, dule is still running and the answer lacking.
leucocytosis, increased alkaline phosphatase (AP) and The value and the optimal timing of resection of
serum bicarbonate were only positive in some of the persistent tumor at the end of chemotherapy in
trials in which they were evaluated, and cannot be sequential protocols or after simultaneous radioche-
integrated in clinical routine to found a treatment motherapy has only been evaluated in one randomized
Prognostic Factors in Lung Cancer 685

trials, and cannot be recommended. It might increase Brundage MD, Feldman-Stewart D, Cosby R et al (2001) Phase
local control, but without influencing overall survival I study of a decision aid for patients with locally advanced
non-small cell lung cancer. J Clin Oncol 19:1326–1335
(Lad et al. 1994). In extensive disease, radiotherapy Brundage MD, Davies D, Mackillop WJ (2002) Prognostic
should not be omitted in treatment responders, because factors in non-small cell lung cancer. Chest 122:1037–1057
local tumor control and median survival can be Buccheri G (1998) Depressive reactions to lung cancer are
improved by additional irradiation (Jeremic et al. common and often followed by a poor outcome. Eur Respir
J 11:173–178
1999). Buccheri G, Ferrigno D (1994) Prognostic factors in lung
cancer: tables and comments. Eur Respir J 7:1350–1364
Buccheri G, Ferrigno D (2004) Prognostic factors. Hematol
References Oncol Clin N Am 18:187–201
Buccheri G, Ferrigno D, Tamburini M (1996) Karnofsky
and ECOG performance status scoring in lung cancer: a
Agarwal M, Brahmanday G, Bajaj SK, Ravikrishnan KP, Wong prospective, longitudinal study of 536 patients from a single
CYO (2010) Revisiting the prognostic value of preoperative institution. Eur J Cancer 32A:1135–1141
18
F-fluoro-2-deoxyglucose (18F-FDG) positron emission Charlson ME, Pompei P, Ales KL, MacKenzie CR (1987)
tomography (PET) in early-stage (I and II) non-small cell A new method of classifying prognostic comorbidity in
lung cancer (NSCLC). Eur J Nucl Med Mol Imaging longitudinal studies: development and validation. J Chronic
37:691–698 Dis 40:373–383
Albain KS, Crowley JJ, Leblanc M, Livingston RB (1990) Choi N, Baumann M, Flentje M et al (2001) Predictive factors
Determinants of improved outcome in small-cell lung in radiotherapy for non-small cell lung cancer: present
cancer: an analysis of the 2,580-patient Southwest Oncol- status. Lung Cancer 31:43–56
ogy Group data base. J Clin Oncol 8:1563–1574 Coate LE, John T, Tsao MS, Shepherd FA (2009) Molecular
Alexander BM, Othus M, Caglar HB, Allen AM (2011) Tumor predictive and prognostic markers in non-small-cell lung
volume is a prognostic factor in non-small-cell lung cancer cancer. Lancet Oncol 10:1001–1010
treated with chemoradiotherapy. Int J Radiat Oncol Biol Emami B, Perez CA (1993) Lung. In: Perez CA (ed) Radiation
Phys 79:1381–1387 oncology. Lippincott, Philadelphia
Al-Saraf N, Gately K, Lucey J, Aziz R, Doddakula K, Wilson Feld R, Borges M, Giner V et al (1994) Prognostic factors in
L, McGovern E, Young V (2008) Clinical implication and non-small cell lung cancer. Lung Cancer 11(suppl):
prognostic significance of standardised uptake value of S19–S23
primary non-small cell lung cancer on positron emission Ferrigno D, Buccheri D, Biggi A (1994) Serum tumour markers
tomography: analysis of 176 cases. Eur J Cardiothor Surg in lung cancer: history, biology and clinical applications.
34:892–897 Eur Respir J 7:186–197
Asmis TR, Ding K, Seymour L, Shepherd FA, Leigh NB, Filosso P, Ruffini E, Asioli S, Giobbe R, Macri L, Bruna MC,
Winton TL, Whitehead M, Spaans JN, Graham BC, Goss Sandri A, Oliaro A (2011) Adensquamous lung carcinomas:
GD (2008) Age and comordidity as independent prognostic a histologic subtype with poor prognosis. Lung Cancer. doi:
factors in the treatment of non small-cell lung cancer. J Clin 10.1016/j.lungcan.2011.01.030
Oncol 26:54–59 Fissler-Eckhoff A (2010) Prognostic factors in histopatholgy of
Bergh JC (1990) Gene amplification in human lung cancer. The lung cancer. In: Heide J, Schmittel A, Kaiser D, Hinkelbein
myc family genes and other proto-oncogenes and growth W (eds) Controversies in the treatment of lung cancer.
factor genes. Am Rev Respir Dis 142:S20–S26 Frontier radiation therapy oncology, vol 42. Karger, Basel,
Birim O, Kappetein AP, van Klaveren RJ, Bogers AJ (2006) pp 1–14
Prognostic factors in non-small cell lung cancer surgery. Fontanini G, De Laurentiis M, Vignati S et al (1998) Evaluation
Eur J Surg Oncol 32:12–23 of epidermal growth factor-related growth factors and
Bonnesen B, Pappot H, Homstav J, Guldhammer Skov B receptors and of neoangiogenesis in completely resected
(2009) Vascular endothelial growth factor A and vascular stage I–IIIA non-small cell lung cancer: amphiregulin and
endothelial growth factor receptor expression in non-small microvessel count are independent prognostic indicators of
cell lung cancer patients: relation to prognosis. Lung Cancer survival. Clin Cancer Res 4:241–249
66:314–318 Ginsberg RJ, Vokes EE, Raben A (1999) Cancer of the lung. In:
Brambilla E, Travis WD, Colby TV et al (2001) The new DeVita VT, Hellman S, Rosenberg SA (eds) Cancer:
World Health Organization classification of lung tumours. principles and practice of oncology. Lippincott-Raven,
Eur Respir J 18:1059–1068 Philadelphia, pp 849–950
Bremnes RM, Sundstrom S, Aasebo U et al (2003) The value of Goldstraw P, Ball D, Jett JR, Le Chevalier T, Lim E, Nicholson
prognostic factors in small cell lung cancer: results from a AG, Shepherd FA (2011) Non-small-cell lung cancer.
randomized multicenter study with minimum 5 year follow- Lancet. doi:10.1016/S0140-6736(10)62101-0
up. Lung Cancer 39:303–313 Graziano SL (1997) Non-small cell lung cancer: clinical value
Bremnes RM, Camps C, Sirera R (2006) Angiogenesis in non- of new biological predictors. Lung Cancer 17:S37
small cell lung cancer: the prognostic impact of neoangio- Harpole DH, Herndone JE, Young WG et al (1995) Stage I non-
genesis and the cytokines VEGF and bFGF in tumours and small cell lung cancer: a multivariate analysis of treatment
blood. Lung Cancer 51:143–158 methods and patterns of recurrence. Cancer 76:787
686 F. B. Zimmermann

Hespanhol V, Queiroga H, Magalhaes A et al (1995) Survival carcinoma treated with radiotherapy. Radiother Oncol
predictors in advanced non-small cell lung cancer. Lung 55:19–25
Cancer 13:253–267 Lebeau B, Chastang C, Schuller MP et al (1995) Chimiothéra-
Hirsch FR, Matthews MJ, Aisner S et al. (1988) Histopatho- pie des cancers bronchiques. Importance prognostique
logic classification of small cell lung cancer. Changing d’une résponse complète (1,280 patients). La Presse Médi-
concepts and terminology. Cancer 62:973–977 cale 24:217–221
Hsu WH, Huang CS, Hsu HS, Huang WJ, Lee HC, Huang BS, Lebeau B, Urban T, Brechot JM et al (1999) A randomized
Huang MH (2007) Preoperative serum carcinoembryonic clinical trial comparing concurrent and alternating thoracic
antigen level is a prognostic factor in women with early irradiation for patients with limited small-cell lung cancer.
non-small-cell lung cancer. Ann Thorax Surg 83:419–424 Cancer 86:1480–1487
Jassem E, Bigda J, Dziadziuszko R et al (2001) Serum p53 Lee JH, Machtay M, Kaiser LR et al (1999) Non-small cell lung
antibodies in small cell lung cancer: the lack of prognostic cancer: prognostic factors in patients treated with surgery and
relevance. Lung cancer 31:17–23 postoperative radiation therapy. Radiology 213:845–852
Jazieh AR, Hussain M, Howington JA et al (2000) Prognostic Lee DS, Kim YS, Kang JH, Lee SN, Kim YK, Ahn MI, Han
factors in patients with surgically resected stages I and II DH, Yoo IR, Wand YP, Park JG, Yoon SC, Jang HS, Choi
non-small cell lung cancer. Ann Thorac Surg 70:1168 BO (2011) Clinical response and prognostic indicators of
Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic (1997) concurrent chemoradiation for non-small cell lung cancer.
Initial versus delayed accelerated hyperfractionated radia- Cancer Res Treat 43:32–41
tion therapy and concurrent chemotherapy in limited Li CT, Marek M, Guclu SZ, Kin Y, Meshref M, Qin S,
small-cell lung cancer: a randomized study. J Clin Oncol Kadziola Z, Krejcy K, Altug S (2011a) Smoking and
15:893–900 prognostic factors in an observational setting in patients
Jeremic B, Shibamoto Y, Nikoloic N et al (1999) Role of with advanced non-small cell lung carcinoma. J Cancer
Radiation Therapy in the combined-modality treatment of 2:52–62
patients with extensive disease small-cell lung cancer: a Li SH, Tian H, Yue WM, Li L, Li WJ, Chen ZT, Hu WS, Zhu YC,
randomized study. J Clin Oncol 17:2092–2099 Qi L (2011b) Overexpression of metastasis-associated pro-
Jeremic B, Classen J, Bamberg M (2002) Radiotherapy alone in tein 1 is significantly correlated with tumor angiogenesis and
technically operable, medically inoperable, early-stage (I/II) poor survival in patients with early-stage non-small cell lung
non-small cell lung cancer. Int J Radiat Oncol Biol Phys cancer. Ann Surg Oncol. doi:10.1245/s10434-010-1510-5
54:119–130 MacKillop WJ (2001) The importance of prognosis in cancer
Jorgensen LG, Osterlind K, Genollá J et al (1996) Serum medicine. In: Gospodarowicz MK, Henson DE, Hutter RBP
neuron-specific enolase (S-NSE) and the prognosis in small- et al (eds) Prognostic factors in cancer. Wiley, Ney York
cell lung cancer (SCLC): a combined multivariate analysis Maione P, Rossi A, Di Maio M, Gridelli C (2009) Tumor-
on data from nine centres. Br J Cancer 74:463–467 related leucocytosis and chemotherapy-induced neutrope-
Kawaguchi T, Takada M, Kubo A, Matsumura A, Fukai S, nia: linked or independent prognostic factors for advanced
Tamura A, Saito R, Kawahara M, Maruyama Y (2010) non-small cell lung cancer? Lung Cancer 66:8–14
Gender, histologoy, and time of diagnosis are important Mandrekar SJ, Schild SE, Hillman SL, Allen KL, Marks RS,
factors for prognosis. J Thorax Oncol 5:1011–1017 Mailliard JA, Krook JE, Maksymiuk AW, Chansky K, Kelly
Kawahara M, Fukuoka M, Saijko N, Nishiwaki Y, Ikegami H, K, Adjei AA, Jett JR (2006) A prognostic model for
Tamura T, Shimoyama M, Suemasu K, Furuse K (1999) advanced stage non small cell lung cancer. Pooled analysis
Prognostic factors and prognostic staging system for small of North Central Cancer Treatment Group trials. Cancer
cell lung cancer. Jpn J Clin Oncol 27:158–165 107:781–792
Komaki R, Fujii T, Perkins P, Ro JY, Allen PK, Mason KA Martins SJ, Takagaki TY, Silva AGP, Gallo CP, Silva FBA,
(1996) Apoptosis and mitosis as prognostic factors in Capelozzi VL (2009) Prognostic relevance of TTF-1 and
pathologically staged N1 nonsmall cell lung cancer. Int J MMP-9 expression in advanced lung adenocarcinoma. Lung
Radiat Oncol Biol Phys 36:601–605 Cancer 64:105–109
Kwiatkowski DJ, Harpole DH Jr, Godleski JH et al. (1998) Matsuoka K, Sumitomo S, Nakashima N, Nakajima D, Misaki
Molecular patholgic substaging in 244 stage I non-small- N (2007) Prognostic value of carcinoembryonic antigen and
cell lung cancer patients: clinical implications. J Clin Oncol Cyfra 21-1 in patients with pathological stage I non-small
16:2468–2477 lung cancer. Eur J Cardio Thor Surg 32:435–439
Lad T, Piaritadosi S, Thomas P et al (1994) A prospective Meert AP, Paesmans M, Martin B et al (2002) The role of
randomized trial to determine the benefit of surgical microvessel density on the survival of patients with lung
resection of residual disease following response of small- cancer: a systematic review of the literature with meta-
cell lung cancer to combination chemotherapy. Chest analysis. Br J Cancer 87:694–701
106:3205–3235 Micke P, Faldum A, Metz T et al (2002) Staging small cell lung
Lamy P, Grenier J, Kramar A, Pujol J (2000) Pro-gastrin- cancer: Veterans Administration Lung Study Group versus
releasing peptide, neuron specific enolase and chromogranin International Association for the Study of Lung Cancer–
A as serum markers of small cell lung cancer. Lung Cancer What limits limited disease? Lung Cancer 37:271–276
29:197–203 Miller MD, Paradis CF, Houck PR, Mazumdar S, Stack JA,
Langendijk H, Aaronson NK, de Jong JM et al (2000) The Rifai AH (1992) Rating chronic medical illness burden in
prognostic impact of quality of life assessed with the geropsychiatric practice and research: application of the
EORCT QLQ-C30 in inoperable non-small cell lung cumulative illness rating scale. Psychiatry Res 41:237–248
Prognostic Factors in Lung Cancer 687

Mizuguchi S, Nishiyama N, Iwata T, Nishida T, Izumi N, Paesmans M, Berghmans T, Dusart M, Garcia C, Hossein-
Tsukioka T, Kiyotoshi Im Uenishi T, Wakasa K, Suehiro S Foucher C, Lafitte JJ, Mascaux C, Meert AP, Roelandts M,
(2007) Serum Sialyl Lewisx and cytokeratin 19 fragment as Scherpereeel A, Munoz VT, Sulier JP (2010) Primary tumor
predictive factors for recurrence in patients with stage I non- standardized uptake value measured on fluorodeoxyglucose
small cell lung cancer. Lung cancer 58:369–375 positron emission tomography is of prognostic value for
Montazeri A, Milroy R, Hole D et al (2001) Quality of life in survival in non-small cell lung cancer. J Thorac Oncol
lung cancer patients: as an important prognostic factor. 5:612–619
Lung Cancer 31:233–240 Palomares MR, Sayre JW, Shekar KC et al (1996) Gender
Mountain CF (1997) Revisions in the International System for influence on weight-loss pattern and survival of non-small
Staging Lung Cancer. Chest 111:1710–1717 cell lung carcinoma patients. Cancer 78:2119–2126
Mountain CF, Lukeman JM, Hammar SP, Chamberlain DW, Pedersen LM, Milman N (1996) Prognostic significance of
Coulson WF, Page DL (1987) Lung cancer classification: thrombocytosis in patients with primary lung cancer. Eur
the relationship of disease extent and cell type to survival in Resp J 9:1826–1830
a clinical trial population. J Surg Oncol 35:147–156 Picardo AL, Diez M, Torres A et al (1996) Analysis of the
Murray N, Coy P, Pater JL et al (1993) Importance of timing for prognostic significance of cytosolic determination of CA
thoracic irradiation in the combined modality treatment of 125, CEA, and SCC in patients with NSCLC. Cancer
limited-stage small-cell lung cancer. J Clin Oncol 11:336–344 77:1066–1072
Murray PV, Soussi T, O’Brian ME et al (2000) Serum p53 Pujol JL, Quantin X, Jacot W et al (2003) Neuroendocrine and
antibodies: predictors of survival in small-cell lung cancer? cytokeratin serum markers as prognostic determinants of
Br J Cancer 83:1418–1424 small cell lung cancer. Lung Cancer 39:131–138
Nakamura H, Taniguchi Y, Miwa K, Adachi Y, Fujioka S, Qian Q, Wand Q, Zhan P, Peng L, Wei SZ, Shi Y, Song Y
Haruki T, Takagi Y, Yurugi Y (2011) Comparison of (2010) The role of matrix metalloproteinase 2 on the
surgical outcomes of thoracoscopic lobectomy, segmentec- survival of patients with non-small cell lung cancer: a
tomy, and wedge resection for clinical stage I non-small cell systematic review with meta-analysis. Cancer Invest
lung cancer. Thorac Cardiov Surg 59:137–141 28:661–669
Naughton MJ, Herndon JE, Shumaker SA et al (2002) The Quejada MI, Albain KS (2004) Prognostic factors in non-small
health-related quality of life and survival of small-cell lung cell lung cancer. In: Sculier JP, Fry WA (eds) Malignant
cancer patients: results of a companion study of CALGB tumors of the lung. Springer, Heidelberg, pp 405–422
9033. Qual Life Res 11:235–248 Quoix E, Purohit A, Faller-Beau M et al (2000) Comparative
Olson JR, Robinson CG, El Naqa I, Creach KM, Drzymala RE, prognostic value of lactate dehydrogenase and neuron-specific
Bloc C, Parikh PJ, Bradley JD (2011) Dose-response for enolase in small-cell lung cancer patients treated with
stereotactic body radiotherapy in early-stage non-small-cell platinum-based chemotherapy. Lung Cancer 30:127–134
lung cancer. Int J Radiat Oncol Biol Phys. doi:10.1016/ Rawson NSB, Peto J (1990) An overview of prognostic factors
j.jrobp.2011.01.038 in small cell lung cancer. A report from the Subcommittee
Osterlind K, Anderson PK (1986) Prognostic factors in small for the Management of Lung Cancer of the United Kingdom
cell lung cancer: a multivariate model base on 778 patients Coordinating Committee on Cancer Research. Br J Cancer
treated with chemotherapy with or without irradiation. 61:597–604
Cancer 46:4189–4194 Ray R, Quantin X, Grenier J, Pujol JL (1998) Predictive factors
Ou S-HI, Zell JA, Ziogas A, Anton-Culver H (2007) Prognostic of tumor response and prognostic factors of survival during
factors for survival of stage I nonsmall cell lung cancer lung cancer chemotherapy. Cancer Detect Prev 22:293–304
patients. Cancer 110:1532–1541 Reinmuth N, Brandt B, Semik M, Kunze WP, Achatzy R,
Ou S-HI, Zell JA, Ziogas A, Anton-Culver H (2008) Low Scheld HH, Broermann P, Berdel WE, Macha HN, Thomas
socioeconomic status is a poor prognostic factor for survival in M (2002) Prognostic impact of Cyfra21-1 ad other serum
stage I nonsmall cell lung cancer and is independent of surgical markers in completely resected non-small cell lung cancer.
treatment, race, and marital status. Cancer 112:2011–2020 Lung Cancer 36:265–270
Ou S-HI, Ziogas A, Zell JA (2009) Asian ethnicity is a Reinmuth N, Thomas M, Meister M, Schnabel PA, Kreuter M
favorable prognostic factor for overall survival in non-small (2010) Current data on predictive markers for anti-
cell lung cancer (SNCLC) and is independent of smoking angiogenic therapy in thoracic tumours. Eur Respir J 36:
status. J Thorac Oncol 4:1083–1093 915–924
Paesmans M (2004) Prognosis of small cell lung cancer. In: Rossi A, Galetta D, Gridelli C (2009) Biological prognostic and
Sculier JP, Fray WA (eds) Malignant tumors of the lung. predictive factors in lung cancer. Oncology 77(Suppl 1):
Springer, Heidelberg, pp 423–432 90–96
Paesmans M, Sculier JP, Libert P et al (1995) Prognostic factors Rygaard K, Vindelov LL, Spang-Thomsen M (1993) Expres-
for survival in advanced non-small-cell lung cancer: sion of myc family oncoproteins in small-cell lung-cancer
univariate and multivariate analyses including recursive cell lines and xenografts. Int J Cancer 54:144–152
partitioning and amalgamation algorithms in 1,052 patients: Sabiston DCJ, Spencer FC (1995) Surgery of the chest. WB
the European Lung Cancer Working Party. J Clin Oncol Saunders, Philadelphia
13:1221–1230 Salgia R, Skarin AT (1998) Molecular abnormalities in lung
Paesmans M, Sculier JP, Lecomte J et al (2000) Prognostic cancer. J Clin Oncol 16:1207–1217
factors for patients with small cell lung cancer. Cancer Saudners M, Dische S, Barrett A et al (1999) Continuous
89:523–533 hyperfractionated accelerated radiotherapy (CHART) versus
688 F. B. Zimmermann

conventional radiotherapy in non-small cell lung cancer: Turrisi AT, Kim K, Blum R et al (1999) Twice-daily compared
mature data from the randomized multicentre trial. Radiother with one-daily thoracic radiotherapy in limited small cell
Oncol 52:137–148 lung cancer treated concurrently with cisplatin and etopo-
Sause W (2001) Nonsurgical management of non-small-cell side. N Engl J Med 340:265–271
lung cancer. Hem Oncol Clin N Am 5:277–289 Wang CY, Huang MS, Huang MH, Lee HC, Hsu HS (2010)
Sculier JP, Chansky K, Crowley JJ, Van Meerbeeck J, Persistently high serum carcinoembryonic antigen levels
Goldstraw P, IASLC International Staging Project (2008) after surgery indicate poor prognosis in patients with stage I
The impact of additional prognostic factors on survival and non-small-cell lung cancer. J Surg Res 163:e45–e50
their relationship with the anatomical extent of disease as Warde P, Payne D (1992) Does thoracic irradiation improve
expressed by the 6th edition of the TNM classification of survival and local control in limited-stage small-cell
malignant tumours and the proposals for the 7th edition. carcinoma of the lung? A meta-analysis. J Clin Oncol
J Thorac Oncol 3:457–466 10:890–895
Sheperd FA, Ginsberg R, Patterson GA et al (1991) Surgical Wheatley-Price P, Le Maitre A, Ding K, Leighl N, Hirsh V,
treatment for limited small-cell lung cancer. J Cardiovasc Seymour L, Bezjak A, Shepherd FA (2010) The influence of
Surg 101:385–393 sex on efficacy, adverse events, quality of life, and delivery
Shepherd FA, Crowley J, Van Houtte P (2007) The IASLC of treatment in National Cancer Institute of Canada Clinical
Lung Cancer Staging Project: proposals regarding the Trials Group Non-small Cell Lunge cancer Chemotherapy
clinical staging of small-cell lung cancer in the forthcoming Trials. J Thorac Oncol 5:640–648
(seventh) edition of the TNM classification for lung cancer. Wigren T, Oksanen H, Kellokumpu-Lehtinen P (1997) A
J Thorac Oncol 2:1067–1077 practical prognostic index for inoperable non-small-cell
Singh S, Parulekar W, Murray N (2005) Influence of sex on lung cancer. J Cancer Res Clin Oncol 123:259–266
toxicity and treatment outcome in small-cell lung cancer. Willner J, Baier K, Caragiani E et al (2002) Dose, volume, and
J Clin Oncol 23:850–856 tumor control predictions in primary radiotherapy of non-
Sobin L, Gospdarowicz M, Wittekind C (eds) (2010) TNM small-cell lung cancer. Int J Radiat Oncol Biol Phys
classification of malignant tumorus, 7th edn. Blackwell, Oxford 52:382–389
Spiegelman D, Maurer LH, Ware JH et al (1989) Prognostic Wolf M, Holle R, Hans K et al (1991) Analysis of prognostic
factors in small-cell carcinoma of the lung: an analysis of factors in 766 patients with small cell lung cancer (SCLC):
1,521 patients. J Clin Oncol 7:344–354 the role of sex as a predictor for survival. Br J Cancer
Stanley KE (1980) Prognostic factors for survival in patients 63:986–992
with inoperable lung cancer. J Natl Cancer Inst 65:25–32 Work E, Nielson OS, Bentzen SM et al (1997) Randomized
Stewart LA, Pignon JP (1995) Chemotherapy in non-small cell study of initial versus late chest irradiation combined with
lung cancer: a meta-analysis using updated data on chemotherapy in limited-stage small-cell lung cancer. J Clin
individual patients from 52 randomised clinical trials. Oncol 15:3030–3037
BMJ 311:899–909 Wu XH, Qian C, Yuan K (2011) Correlations of hypoxia-
Subramanian J, Simon R (2010) What should physicians look inducible factor-1a/hypoxia-inducible factor-2a expression
for in evaluating prognostic gene-expression signatures? with angiogenesis factors expression and prognosis in non-
Nat Rev Clin Oncol 7:327–334 small cell lung cancer. Chin Med J 124:11–18
Sunaga N, Tsuchiya S, Minato K et al (1999) Serum pro- Würschmidt F, Bünemann H, Heilmann HP (1995) Small cell
gastrin-releasing peptide is a useful marker for treatment lung cancer with and without vena cava syndrome: a
monitoring and survival in small-cell lung cancer. Oncology multivariate analysis of prognostic factors in 408 cases. Int J
57:143–148 Radiat Oncol Biol Phys 33:77–82
Tai P, Tonita J, Yu E, Skarsgard D (2003) Twenty-year follow- Yip D, Harper PG (2000) Predictive and prognostic factors
up study of long-term serviva of limited-stage small-cell in small cell lung cancer: current status. Lung Cancer 28:
lung cancer and overview of prognostic and treatment 173–185
factors. Int J Radiat Oncol Biol Phys 56:626–633 Zhan P, Wnag J, Lv XJ, Wang Q, Qiu LX, Lin XQ, YuYSong
Takada M, Fukuoka M, Kawahara M et al (2002) Phase III LK, Song Y (2009) Prognostic value of vascular endothelial
study of concurrent versus sequential thoracic radiotherapy growth factor expression in patients with lung cancer.
in combination with cisplatin and etoposide for limited- J Thorac Oncol 4:1094–1103
stage small cell lung cancer: results of the Japan Clinical Zimmermann FB, Bamberg M, Molls M, Jeremic B (2003)
Oncology Group study 9104. J Clin Oncol 20:3054–3060 Radiation therapy alone in early stage non-small-cell lung
Takigawa N, Segawa Y, Okahara M et al (1996) Prognostic cancer. Semin Surg Oncol 21:91–97
factors for patients with advanced non-small cell lung cancer: Zimmermann F, Wulf J, Lax I, Nagata Y, Timmermann RD,
univariate and multivariate analyses including recursive Stojkivski I, Jeremic B (2010) Stereotactic body radiation
partitioning and amalgamation. Lung Cancer 15:67–77 therapy for early non-small cell lung cancer. In: Heide J,
Tamura M, Ueoka H, Kiura K et al (1998) Prognostic factors of Schmittel A, Kaiser D, Hinkelbein W (eds) Controversies in
small-cell lung cancer in Okayama Lung Cancer Study the treatment of lung cancer. Frontier radiation therapy
Group Trials. Acta Med Okayama 52:105–111 oncology, vol 42. Karger, Basel, pp 94–114
Intensity-Modulated Radiation Therapy
and Volumetric-Modulated Arc Therapy
for Lung Cancer
Inga S. Grills and Victor S. Mangona

Contents 6 Outcomes .................................................................. 705


7 Conclusion ................................................................ 709
1 Introduction.............................................................. 692 References.......................................................................... 709
2 Technical Aspects .................................................... 692
2.1 Beam Modification.................................................... 692
2.2 Three-Dimensional Conformal Radiation Therapy... 693 Abstract
2.3 Intensity-Modulated Radiation Therapy ................... 693 Per 2010 estimates by the American Cancer
2.4 Intensity-Modulated Arc Therapy............................. 694
2.5 Volumetric-Modulated Arc Therapy......................... 695
Society, lung cancer has both the highest incidence
and mortality of all malignancies in the United
3 Potential Advantages of
States. Overall, outcomes, though improving,
Intensity-Modulated Radiation Therapy
and Volumetric-Modulated Arc Therapy: remain poor, and radiation therapy (RT) is an
Dose-Escalation and Toxicity Reduction .............. 696 important mainstay of locoregional therapy. The
3.1 Intensity-Modulated Radiation Therapy versus technical challenges of delivering biologically
Three-Dimensional Conformal Radiotherapy........... 696
effective doses of RT capable of achieving
3.2 Intensity-Modulated Radiation Therapy Planning
Studies........................................................................ 696 adequate local control are many and relate to
3.3 Toxicity and Intensity-Modulated Radiation target definition, respiratory tumor motion, tissue
Therapy ...................................................................... 696 heterogeneities and normal tissue tolerance.
3.4 Volumetric-Modulated Arc Therapy Experience ..... 700
Advancements over standard two-dimensional
4 Treatment Planning................................................. 700 RT, including three-dimensional conformal radia-
4.1 Simulation .................................................................. 701 tion therapy (3D-CRT), intensity-modulated radi-
4.2 Target Volumes (Gross Tumor Volume, Clinical
Target Volume, Planning Target Volume)............... 702 ation therapy (IMRT), and more recently
4.3 Inverse Planning ........................................................ 702 volumetrically-modulated arc therapy (VMAT),
5 Targeting and Verification ..................................... 703
in addition to four-dimensional (4D) CT simula-
5.1 Image-Guided Radiation Therapy............................. 703 tion and planning techniques, using biological
5.2 Biological Targeting with Positron Emission targeting via positron emission tomography (PET)
Tomography............................................................... 704 and 2 and 3-D image-guided delivery methods
5.3 Treatment Verification .............................................. 705
have aided the path toward achieving radiation
dose escalation while concurrently sparing organs
at risk (OAR) and reducing target miss. IMRT
planning studies have shown improvements over
I. S. Grills (&)  V. S. Mangona 3D-CRT with respect to tumor dose escalation and
Department of Radiation Oncology, OAR dose, particularly for locally-advanced dis-
William Beaumont Hospital,
ease. This, however, is potentially at the cost of
3601 West Thirteen Mile Road,
Royal Oak, MI 48073-6769, USA longer treatment times, monitor units, and volume
e-mail: igrills@beaumont.edu of lung receiving low dose, with some concern for

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 691
DOI: 10.1007/174_2011_283,  Springer-Verlag Berlin Heidelberg 2011
692 I. S. Grills and V. S. Mangona

higher pneumonitis rates. Commercial planning Michigan group reached doses up to 102.9 Gy for
software and quality assurance measures now small solitary tumors (Rengan et al. 2004). Across
allow for dynamically delivered VMAT, which dose-escalation studies, however, the maximum tol-
similarly may achieve higher tumor dose and lower erated dose for standard fractionated RT, without
OAR dose while concurrently reducing treatment chemotherapy, has been approximately 83–84 Gy.
times and monitor units and increasing target Exceedingly high-biological effective doses (BEDs)
conformality. Though IMRT and VMAT planning are now routinely administered for the treatment of
studies and short-term, single-institution clinical data stage I NSCLC by way of stereotactic body radiation
are encouraging, long-term, multi-institutional stud- therapy (SBRT) where a BED of [94–105 Gy has
ies comparing these techniques to 2D or 3D-CRT in shown higher rates of local control (Grills et al.
terms of locoregional control, survival, and quality of 2010a; McGee et al. 2010; Onishi et al. 2010; Wulf
life are lacking but should be supported. et al. 2005). In RTOG 0236, SBRT yielded a 3-year
LC rate of 98% (Timmerman et al. 2010), and
emerging data have shown SBRT to have comparable
results to sublobar (e.g. wedge) resection in terms of
1 Introduction local and regional recurrence (Grills et al. 2010b;
Welsh et al. 2010).
In the US, 2010 estimates by the American Cancer Even with the advent of three-dimensional confor-
Society suggest that lung cancer has both the highest mal radiotherapy (3D-CRT), escalating to tumoricidal
cancer incidence: 222,000 new cases per year (more doses for larger tumors is highly constrained by the
than both breast and prostate cancer) and highest potential for toxicity to the spinal cord, esophagus
yearly cancer mortality: 157,000 deaths per year, (esophagitis/dysphagia), and normal lung (pneumoni-
nearly as many as the next four leading causes com- tis). Furthermore, the amplified risk of ‘‘geographic
bined: colon, breast, pancreas, and prostate (American miss’’ due to respiratory tumor motion is a challenge
Cancer Society: Cancer Facts and Figures 2010). virtually unique to intrathoracic malignancies.
In patients with inoperable non-small-cell lung Particularly in locally-advanced disease, the advent of
cancer (NSCLC), radiation therapy remains the pri- intensity-modulated radiation therapy (IMRT) increa-
mary curative modality for both early-stage and ses the potential for dose-escalation while sparing
locally-advanced disease, despite chemotherapeutic organs at risk (OARs), but requires advanced technol-
advances (Arriagada et al. 1991; Furuse et al. 1999; ogies to account for respiratory tumor motion such as
Pisters 2000). Delivery of radiation therapy (RT) for four-dimensional computed tomography (4D-CT) and
intrathoracic malignancies is technically challenging, is ideally administered using online 3D-image-guid-
and conventional radiotherapy doses and techniques ance for proper targeting and margin reduction.
have yielded unsatisfactory results. In early-stage In this chapter, we discuss technological advances
disease, local failure occurs in 60–70% of patients beyond 3D-CRT, particularly IMRT, intensity-modu-
with 2-year survival under 40% using standard- lated arc therapy (IMAT), and volumetric-modulated
fractionated RT (Armstrong and Minsky 1989; arc therapy (VMAT) and treatment planning, toxicity,
Dosoretz et al. 1996; Kaskowitz et al. 1993). and clinical outcomes associated with these modalities.
In locally-advanced disease, 3-year local control (LC)
is approximately 20% with median survival under
18 months (Curran et al. 2003; Ataman et al. 2001). 2 Technical Aspects
Studies from the University of Michigan, radiation
therapy oncology group (RTOG), and Memorial 2.1 Beam Modification
Sloan-Kettering Cancer Center (MSKCC) have
shown improved local control with dose-escalation Upon exiting a treatment machine, beam modification
(Hayman et al. 2001; Narayan et al. 2004; Bradley occurs by many methods for different purposes.
et al. 2005; Rosenzweig et al. 2005). In large tumors Custom blocks (e.g. historically lead or cerrobend)
(C100 ml), Rengan et al. showed a 36% decrease in shape beams for target conformality a job now most
local failure with a 10 Gy increase in dose, and the commonly replaced by multi-leaf collimators (MLCs)
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 693

Fig. 1 A close-up photo of MLC leaves looking up into the


collimator head (Copyright  2010, Varian Systems, Inc. All
rights reserved.)

(Figs. 1, 2), which make IMRT possible. Flattening


filters improve dose homogeneity from central axis to Fig. 2 An MLC portal outline (beams-eye view) as shown on
block edge. Wedges confer a linear gradient of dose a computer display. This MLC is a tertiary system. The position
of the secondary jaws is shown. There are locations where the
fluence across a beam in a single direction, whereas leaves overlap the desired treatment outline and other spots
physical compensators manipulate dose intensity in where they under lap producing a ‘‘scalloped’’ contour. The leaf
two dimensions. Irradiation with wedges and/or width is 1.0 cm projected to isocenter. Varian SHAPER
compensators is IMRT in its simplest form, radiation program software (Copyright  2010, Varian Medical Systems,
Inc. All rights reserved.)
with heterogeneous dose fluence across the beam.
Although no universally-accepted definition of or a single MLC portal) is allowed for each beam
IMRT exists, Bortfield described IMRT in practice as ‘‘a position. If, for example, three different blocks (beam
radiation treatment technique with multiple beams, in shapes) were used at a single beam angle, the integral
which at least some of the beams are intensity-modu- dose fluence from the three beams would create a map
lated and intentionally deliver a non-uniform intensity to of variable dose fluence (Fig. 3) (McDermott and
the target. The desired dose distribution in the target is Orton 2010), resulting in an individual IMRT beam.
achieved after superimposing such beams from different
directions. The additional degrees of freedom are
utilized to achieve a better target dose conformity and/or 2.3 Intensity-Modulated Radiation
better sparing of critical structures (Bortfield 2006).’’ Therapy

2.2 Three-Dimensional Conformal In practice, manually cutting blocks for multiple


Radiation Therapy beam segments and angles, then—in turn—manually
placing each block for each segment would require
Three-dimensional treatment planning became possi- great time, manual effort, and substantial potential for
ble with the advent of computed tomography in the error. The advent of computer-generated and manip-
1970s and the routine availability of computers in the ulated MLCs (Figs. 1, 2) is what made IMRT1
1980s. With 3D-planning, target volumes and organs
at risk (OARs) can be well-defined; and beam shapes, 1
For the purposes of this chapter, the abbreviation ‘‘IMRT’’ by
directions, and energy levels can be appropriately default will refer only to fixed-field IMRT, whereby intensity-
selected to give rise to increased conformity, modulated beams are delivered from multiple discrete, fixed
decreased margins, and target dose-escalation with angles (using segmental or dynamic MLCs) without any gantry
rotation during beam-on time, thus excluding techniques such
increased sparing of normal tissue. In practice,
as tomotherapy, IMAT, VMAT, etc. The unabbreviated term
3D-CRT implies that for each given beam angle, only ‘‘intensity-modulated radiation therapy’’ may, however, confer
a single beam shape modifier (e.g. a cerrobend block a broader connotation.
694 I. S. Grills and V. S. Mangona

Fig. 3 Left intensity map of an IMRT beam superimposed on a IMRT beam intensity (left) (From McDermott and Orton, color
patient digital reconstructed radiograph (DRR). Right radiation- plate 18, The physics and technology of radiation therapy, 
induced epilation on the patient’s scalp from corresponding 2010)

Fig. 4 An intensity map intensities delivered through multiple beam angles


showing an IMRT beam with
allows dose-escalation to target volumes and sub-
1 cm 9 1 cm beamlets with
ten different greyscale volumes (e.g. simultaneous integrated boost), organ
intensity levels (white less sparing, and ultimately, ‘‘dose painting’’ (Figs. 6, 7)
intense, dark more intense) superior to 3D-CRT (Brahme 1988).
(From McDermott and Orton,
color plate 17, The physics
and technology of radiation
therapy,  2010) 2.4 Intensity-Modulated Arc Therapy

Yu et al. at William Beaumont Hospital first intro-


practical and logistically feasible, allowing large
duced intensity-modulated arc therapy2 (IMAT) in
numbers of ‘‘segments’’ (‘‘apertures’’). In segmental
1995. This was a rotational radiation therapy delivery
IMRT, the most common form, the beam turns off
technique in which the field shape changed dynami-
whenever the MLCs are in motion and is on only
cally as the linear accelerator gantry rotated.
when the MLCs are stationary, forming a static portal.
Gantry rotational speed and dose rate were constant
The initial beam segment for a given beam position
may be the ‘‘open-field’’ (e.g. which might be used for
a 3D-conformal plan). An electronic portal imaging 2
Notably, distinctions for IMAT and VMAT are not univer-
device (EPID) routinely takes a beam’s-eye-view sally agreed upon. The term ‘‘arc therapy’’ will refer both to
image through this segment for target verification. IMAT and VMAT. As the term ‘‘VMAT’’ corresponds to
Multiple ‘‘subportal’’ beams, with variable weights technological advances of IMAT, Yu et al. refer to VMAT
expressly as IMAT. Further, VMAT technology has been
are used to modify the initially-delivered open-field trademarked with Elekta (VMATTM), Varian (RapidArcTM),
beam. The resultant ‘‘IMRT beam’’ represents the and Philips (SmartArcTM) and has also been referred to as
summation of dose from all beam segments (the open- ‘‘arc-modulated radiation therapy’’ (AMRT). Here forward, the
and sub-portals). The IMRT beam is therefore a series terms ‘‘volumetric-modulated arc therapy’’ and ‘‘VMAT’’ will
refer generically to the advanced IMAT technology inclusive of
of ‘‘beamlets,’’ with each beamlet providing a variable gantry velocity and variable dose rate and exclusive of
different discrete intensity. At risk of oversimplifica- arc therapy delivered with uniform dose rate and uniform
tion, whereas a 3D-CRT beam represents a true gantry velocity, which will be referred to as intensity-modu-
‘‘black and white’’ image (e.g. a beamlet is either lated arc therapy or ‘‘IMAT.’’ Furthermore, tomotherapy will
be considered its own modality (not to be incorporated by
blocked or unblocked), the IMRT beam permits many default with the terms ‘‘IMRT,’’ ‘‘arc therapy,’’ ‘‘IMAT,’’
shades of gray (Figs. 4, 5). Such heterogeneous beam or ‘‘VMAT.’’
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 695

Fig. 7 The dose distribution in a geometric phantom with


three targets. The targets (red) are cylinders with axes
perpendicular to the page. A seven-field IMRT plan revealed
high conformity with only a single isocenter (intersection of
crosshairs). The maximum dose within the target is 115% the
Fig. 5 Comparison of conventional RT to IMRT. Three beams prescribed dose (From McDermott and Orton, color plate 20,
are used to treat a target with an irregular shape. In the The physics and technology of radiation therapy,  2010)
conventional treatment (upper), the jaws are used to shape
the beams to correspond to the beam’s-eye-view shape of the
target. The resulting dose distribution is shown as homogenous
shading (upper-right). With IMRT (lower), the beam is (Yu 1995; Yu et al. 1995). IMAT did not achieve
modulated in an attempt to reduce the dose to the ‘‘nooks and widespread use for several reasons: the arc sequenc-
crannies’’ representative of normal tissue. The resulting heter- ing was a lengthy process; accurate delivery was
ogeneous dose distribution (lower-right) shows dose reduction
inefficient with complex fields—often multiple
in these regions (From McDermott and Orton, Fig. 20.3, The
physics and technology of radiation therapy,  2010) superimposed arcs were necessary—and available
computer power and memory at the time were inad-
equate for dose calculations at a fine resolution;
further, support from linear accelerator control soft-
ware was lacking (Otto 2008; Ramsey et al. 2001).
Achievable dose distributions, however, were con-
sidered comparable to helical tomotherapy while
using a standard linear accelerator (Cao et al. 2007).

2.5 Volumetric-Modulated Arc Therapy

In pursuit of single-arc IMAT plans with ideal plan


quality, Otto developed an algorithm that allowed for
Fig. 6 The dose distribution in a geometric phantom (square a variable dose rate during arc delivery referred to as
slabs in virtual water) illustrating the power of IMRT. Seven-
gantry angles and five intensity levels were used. The target VMAT2 (Otto 2008). Industrial advances allowed the
is the red annulus encompassing a cylindrical organ at risk development of VMAT as a more dynamic radiation
(e.g. spinal cord). The 100% isodose line wraps tightly around delivery method. Similar to IMAT, the beam is on
both the inside and outside of the annulus giving high during rotation of a standard linear accelerator gantry.
conformity. Dose within the target, however, is quite hetero-
geneous with maximum dose of 148% the prescribed dose However, the dose rate, rotational gantry speed, MLC
(From McDermott and Orton, color plate 19, The physics and position, and collimator angle, are all dynamically-
technology of radiation therapy,  2010) modulated during RT delivery. Thus highly confor-
mal treatment plans could be delivered in single to
throughout treatment delivery. Multiple arcs with multiple arcs. With VMAT, radiation treatment times
varying field shapes created an intensity-modulated can average only 1.5–3 min/2 Gy fraction, substan-
arc. For treatment planning, arcs were planned using tially lower than required for similarly complex
set control points: IMRT beams at incremental angles IMRT plans (Otto 2008). Though VMAT may be
696 I. S. Grills and V. S. Mangona

planned as a near-to-full single rotation (e.g. (MLD) by 2 Gy and amount of lung receiving 20 Gy
340–360) (Otto 2008; Scorsetti et al. 2010; Wolff (lung V20) by 8%. Grills et al. (2003) performed a
et al. 2009; Guckenberger et al. 2009; Clark et al. systematic planning analysis comparing IMRT to two
2010; Ong et al. 2010), it can also be delivered as a 3D-CRT techniques: (1) optimized (numerous-field)
partial arc (e.g. 180) (Matuszak et al. 2010; McGrath 3D-CRT and (2) traditional limited-field (e.g. 2–3
et al. 2010), dual (back-and-forth) partial arcs (Holt beams) 3D-CRT. This study showed significant ben-
et al. 2011), a full ? partial arc (Wolff et al. 2009), efit with IMRT, particularly in node-positive patients
and multiple arcs, both coplanar and non-coplanar and those with target volumes close to the esophagus.
(Guckenberger et al. 2009; Clark et al. 2010). The use With higher mean target doses secondary to IMRT
of multiple arcs can provide further intensity modu- dose heterogeneity, tumor control probability (TCP)
lation or allow for more optimal normal tissue sparing was increased by 7–8%, whereas lung V20, lung
and avoid spreading low dose unnecessarily to other NTCP (normal tissue complication probability), and
areas of the body. The development of IMAT and its esophagus NTCP were reduced by approximately 15,
progression to VMAT are well described in a review 30, and 55%, respectively. For GTVs within 1.5 cm
by Yu and Tang (2011). of the esophagus, IMRT reduced esophagus V50 by
40%. IMRT plans allowed for dose-escalation by
25–30% beyond that of comparably safe 3D-CRT
3 Potential Advantages of plans. A comparison of 3D-CRT to IMRT with
Intensity-Modulated Radiation respect to critical organ dose for various planning
Therapy and Volumetric- studies is included in Table 1.
Modulated Arc Therapy:
Dose-Escalation and Toxicity
Reduction 3.3 Toxicity and Intensity-Modulated
Radiation Therapy
3.1 Intensity-Modulated Radiation
Therapy versus Three-Dimensional Sura et al. (2008) published the largest known toxicity
Conformal Radiotherapy and outcome study of patients with NSCLC treated
exclusively with IMRT. This retrospective analysis
As compared to 3D-CRT, IMRT has many inherent included 55 patients with inoperable stage I–II
potentially advantageous qualities. With heterogeneous (n = 16) and III (n = 39) NSCLC from 2001 to 2005
beam intensities, tumor heterogeneity can be increased, treated with 60 Gy in 2 Gy fractions at MSKCC.
allowing a higher maximum dose in the target as well as Toxicity was scored using a modified RTOG toxicity
sharper dose fall-off close to critical OARs leading to scoring system (Cox et al. 1995). The overall crude
normal tissue sparing (lung, esophagus, spinal cord, rate of any pulmonary toxicity was 13%. Acute and
etc.). Schwarz et al. (2005) showed that for large con- late (C4 months after RT start) toxicities are seen in
cave tumors treated with IMRT the average dose Table 2. Acute grade 1–2 esophagitis was common
increase was as high as 35% compared to 3D-CRT. The (69%), but grade 3 rare (4%). All late esophageal
ability to spare cylindrical structures is well illustrated in toxicity was grade 1–2 and occurred in 22% of
Fig. 6. Figure 7 illustrates the conformity to multiple patients. Grade 2 or higher acute pulmonary toxicity
targets while using a single isocenter, a common sce- rates were 18% grade 2, 11% grade 3, and 0% grade
nario in LA-NSCLC, where tumor and nodal gross 4–5. One patient had late grade 3 pulmonary toxicity,
tumor volumes (GTVs) may be disjointed. and one died (grade 5 toxicity) secondary to treat-
ment-related pneumonitis (TRP) (Sura et al. 2008).

3.2 Intensity-Modulated Radiation 3.3.1 Sparing the Esophagus


Therapy Planning Studies The predominant increase in acute toxicity with
concurrent over sequential chemoradiotherapy for
In comparison to 3D-CRT, Liu et al. (2004) showed locally advanced NSCLC is esophagitis, which can
that nine-field IMRT plans decreased mean lung dose ultimately become dose-limiting (Schwarz et al.
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 697

Table 1 Comparison of median/mean dose to organs at risk: 3D-CRT versus IMRT and VMAT
Organ Study N Patients Parameters 3D- IMRT VMAT Difference P
CRT
Spinal Bedford and 10 NR Dmax 10 Gy – 9 Gy -0.5 Gy NS
cord Warrington (2009)
Grills et al. (2003)a 9 N0 Dmax+3 mm 24 Gy 30 Gy – +5.6 Gy –
Grills et al. (2003)a 9 N2–N3 Dmax+3 mm 41 Gy 41 Gy – -0.6 Gy –
b
Lievens et al. (2011) 35 N2–N3 Dmax 47 Gy 45 Gy – -2.5 Gy –
Ong et al. (2010) 18 I Dmax 8 Gy – 11 Gy +2.9 Gy 0.014
Lungs Lievens et al. (2011) 35 N2–N3 V5 55% 45% – -10% \0.0001
Liu et al. (2004) 10 I–IIIB V5 NR NR – +8.0% 0.007
c c
McGrath et al. (2010) 21 I V5 NR – NR 4.2% RR 0.03
Murshed et al. (2004) 41 III–IV V5 52% 59% – +7% NS
d
Ong et al. (2010) 18 I V5 18% – 18% +0.2% NS
Bedford and 10 NR V10 40% – 34% –5.6% 0.03
Warrington (2009)
Liu et al. (2004) 10 I–IIIB V10 NR NR – –1.6% NS
c c
McGrath et al. (2010) 21 I V10 NR – NR 2.6% RR 0.01
Murshed et al. (2004) 41 III–IV V10 45% 38% – –7% \0.0001
McGrath et al. (2010)c 21 I V12.5 NR – NR 3.2% RRc 0.01
Bedford and 10 NR V20 21% – 24% +2.6% 0.02
Warrington (2009)
Grills et al. (2003)a 9 N0 V20 22% 19% – -2.8% –
a
Grills et al. (2003) 9 N2–N3 V20 29% 26% – -3.2% –
Lievens et al. (2011) 35 N2–N3 V20 28% 27% – -1.2% 0.06
Liu et al. (2004) 10 I–IIIB V20 NR NR – -8.0% 0.005
McGrath et al. (2010)c 21 I V20 NR – NR 4.5% RRc 0.02
Murshed et al. (2004) 41 III–IV V20 35% 25% – -10% \0.0001
Ong et al. (2010)d 18 I V20 4.9% – 5.4% +0.5% 0.025
Liu et al. (2004) 10 I–IIIB V30 NR NR – -8.9% 0.005
Liu et al. (2004) 10 I–IIIB Veff NR NR – -9.0% 0.005
Murshed et al. (2004) 41 III–IV Veff 71 Gy 58 Gy – -13 Gy \0.0001
Bedford and 10 NR MLD 14 Gy – 14 Gy +0.1 Gy NS
Warrington (2009)
Grills et al. (2003)a 9 N0 MLD 13 Gy 12 Gy – -1.8 Gy –
Grills et al. (2003)a 9 N2–N3 MLD 18 Gy 16 Gy – -1.8 Gy –
Liu et al. (2004) 10 I–IIIB MLD NR NR – -2.0 Gy 0.005
Murshed et al. (2004) 41 III–IV MLD 19 Gy 17 Gy – -2 Gy \0.0001
Lievens et al. (2011) 35 N2–N3 MLD 16 Gy 16 Gy – +0.1 Gy NS
Murshed et al. (2004) 41 III–IV Integral lung 19 Gy 16 Gy – -3 Gy \0.0001
dose
Grills et al. (2003)a 9 N0 NTCP 12% 11% – +1.1% –
a
Grills et al. (2003) 9 N2–N3 NTCP 21% 17% – -3.6% –
(continued)
698 I. S. Grills and V. S. Mangona

Table 1 (continued)
Organ Study N Patients Parameters 3D- IMRT VMAT Difference P
CRT
Esophagus Grills et al. (2003)a 9 N0 V50 5% 6% – +0.6% –
a
Grills et al. (2003) 9 N2–N3 V50 26% 19% – -7.6% –
Grills et al. (2003)a 9 N0 Mean dose 12 Gy 12 Gy – -0.2 Gy –
Grills et al. (2003)a 9 N2–N3 Mean dose 27 Gy 24 Gy – -2.9 Gy –
Lievens et al. (2011) 35 N2–N3 Dmax 93 Gy 79 Gy – -14 Gy \0.0001
Grills et al. (2003)a 9 N0 Dmax+3 mm 46 Gy 43 Gy – -3.4 Gy –
Grills et al. (2003)a 9 N2–N3 Dmax+3 mm 76 Gy 74 Gy – -1.6 Gy –
a
Grills 2003(2003) 9 N0 NTCP 5.2% 2.2% – -3.0% –
Grills et al. (2003)a 9 N2–N3 NTCP 41% 19% – -22% –
Heart Bedford and 10 NR Mean dose 9.9 Gy – 9.4 Gy NS –
Warrington (2009)
Grills et al. (2003)a 9 N0 D33 8.9 Gy 9.3 Gy – +0.4 Gy –
a
Grills et al. (2003) 9 N2–N3 D33 19 Gy 20 Gy – +0.8 Gy –
Grills et al. (2003)a 9 N0 D67 5.2 Gy 4.4 Gy – -0.8 Gy –
a
Grills et al. (2003) 9 N2–N3 D67 7 Gy 8 Gy – +1.5 Gy –
Grills et al. (2003)a 9 N0 D100 0.3 Gy 0.6 Gy – +0.3 Gy –
Grills et al. (2003)a 9 N2–N3 D100 0.9 Gy 1.6 Gy – +0.7 Gy –
Grills et al. (2003)a 9 N0 NTCP 0.1% 0% – -0.1% –
a
Grills et al. (2003) 9 N2–N3 NTCP 20% 11% – -9% –
3D-CRT 3D-conformal radiation therapy, IMRT intensity-modulated radiation therapy, VMAT volumetric-modulated radiation
therapy, Difference IMRT-3D-CRT or VMAT-3D-CRT, n number of patients, Dmax maximum dose, Veff = biologically effective
volume, +3 mm = volume of organ ? 3 mm expansion, NR not reported data, NS not statistically significant, I, II, III,
IV = AJCC stage, N nodal staging
a
Both plans with equivalent tumor control probability for PTVGTV. Lung dose calculations exclude gross tumor volume (GTV)
b
Liu 2004-both plans with dose-escalation with calculations using pencil-beam algorithm
c
Only VMAT relative (not absolute) reduction reported in this study. All variables were lower with VMAT in comparison
to 3D-CRT
d
Lung volume excludes planning treatment volume (PTV)

Table 2 Rate of early (\4 mos) and late (C4 mos) reactions in patients treated with IMRT (adapted from Sura et al. 2008)
Grade 0 (%) Grade 1 (%) Grade 2 (%) Grade 3 (%) Grades 4–5 (%)
Esophageal Early 27 47 22 4 –
Late 78 16 6 – –
Pulmonary Early 4 67 18 11 –
Late 23 57 16 2 –
Fatigue Early 18 49 31 – –
Nausea Early 73 22 6 – –
Cardiac Early 98 – – 2 –
Skin Early 45 44 7 4 –
Toxicity was scored using a modified RTOG toxicity scoring system (Cox et al. 1995)
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 699

2005; Liu et al. 2004; Grills et al. 2003; Chapet et al. V10 and V13 to be most predictive of TRP (V5 was not
2005; Murshed et al. 2004). With twice-daily hyper- analyzed). Beaumont data further note V5 (cutoff
fractionated regimens, severe acute esophageal tox- 50%) and V10 (cutoff 45%) as the strongest predictors
icity can be as high as 40 to 60% (Werner-Wasik et al. for grade C3 pneumonitis (Shaitelman et al. 2009).
1999; Choy et al. 1998; Lau et al. 2001). In NSCLC Wang’s multivariate analysis of 223 patients treated
patients treated with either 3D-CRT or IMRT, with concurrent chemo radiation further support V5 as
Shaitelman et al. (2009) found esophagitis to be lin- the strongest predictor of TRP (cutoff 42%) (Wang
early related to esophagus V10/20/30/40/50/55, mean et al. 2006).
dose, and maximum dose. The strongest predictors of Yom et al. evaluated such parameters in patients
grade C2 esophageal toxicity were esophagus treated with IMRT (n = 68) as well as 3D-CRT
V10 [65% and esophagus Dmax (maximum dos- (n = 222) at MD Anderson, particularly predictors of
e)[55 Gy. Multiple studies have shown the potential grade C3 TRP with LA-NSCLC receiving concurrent
for esophageal dose reduction with IMRT over chemotherapy. This study revealed a fourfold
3D-CRT (Liu et al. 2004; Grills et al. 2003; Murshed decrease in TRP with IMRT over 3D-CRT (8 vs.
et al. 2004; Lievens et al. 2011). One of the planning 32%, P = 0.002). IMRT plans had higher V5 (63 vs.
challenges in advanced stage NSCLC is the potential 57%, P = 0.011), similar V10 (48 vs. 49%, P = 0.87),
for overlap of the planning target volume for tumor or and decreased V15, V20…, V65 (P \ 0.05). The
nodes with the esophagus. Chapet et al. (2005, 2006) 12-month incidence of TRP for IMRT-treated patients
have shown that by relaxing homogeneity constraints was significantly less with V5 B 70% (2 vs. 21%,
and using equivalent uniform dose (EUD) calcula- P = 0.02), suggesting a potential V5 threshold of
tions, the planning target volume (PTV) excluding the 70 Gy (Yom et al. 2007). Interestingly, however, the
esophagus can be dosed to higher levels with potential overall rates of TRP were lower with IMRT than with
for better tumor control without increasing esophagus 3D-CRT despite higher V5 in the IMRT group, sug-
NTCP. Similarly, IMRT has been used to boost PET- gesting that other factors may play a significant role in
positive disease using simultaneous integrated boost determining risk for TRP in irradiated patients.
(SIB) to doses 22% higher without significant change Shi et al. (2010) further expounded upon Yom’s
in toxicity (Rebueno and Welsh 2009). work, performing univariate and multivariate analyses
on 94 consecutive patients (11 who developed TRP)
3.3.2 Pneumonitis: Worse with Intensity- treated with concurrent chemotherapy and IMRT.
Modulated Radiation Therapy? Univariate analysis showed that COPD, FEV1, MLD,
Although IMRT has numerous potential advantages lung NTCP, and lung V5–60 were all related to TRP.
over 3D-CRT for NSCLC, IMRT plans require more Multivariate analysis revealed lung NTCP (cutoff
monitor units (MUs) per treatment and thus have risks 4.2%) and V10 (cutoff 50%) most significantly
associated with increased total body exposure. Longer associated with TRP.
treatment times may further increase the potential for
intrafraction variation. In the treatment of intratho- 3.3.3 Reducing Low Dose to Large Volume
racic malignancies, a significant concern is the With the increased low dose to larger lung volumes
potential for increased lung toxicity—in comparison with IMRT, attempts have been made to reduce low-
to 3D-CRT—from spreading low dose throughout a dose volumes, such as ‘‘hybrid-IMRT’’—whereby
larger volume of normal lung. Reduced diffusing static and IMRT treatments are used to decrease
capacity of carbon monoxide (DLCO) has been seen contralateral lung V5, V13, and V20 (Mayo et al. 2008).
in volumes of lung receiving C13 Gy (Gopal et al. Restricting IMRT plans to five or fewer beams has
2003). Many studies have also consistently reported also been associated with reduction of low-dose vol-
the correlation between lung dose and TRP (Willner umes (e.g. V5 and V10) (Liu et al. 2004). Three, five,
et al. 2003; Fay et al. 2005; Schallenkamp et al. 2007; and seven-beam arrangements have been shown to
Shi et al. 2010; Yom et al. 2007; Wang et al. 2006). In have similar MLD, V13, V20, and V30 (Chapet et al.
a 3D-CRT dose-escalation study, high-grade TRP was 2006). A similar study, using beam angle optimiza-
related to low-dose lung RT volumes: V5, V10, and V13 tion to balance beam angle preference for both
(Yorke et al. 2005). Schallenkamp et al. (2007) found tumor and lung, showed that five- and seven-beam
700 I. S. Grills and V. S. Mangona

Fig. 8 Left ten-field non-coplanar beam 3D-CRT plan for SBRT. Right corresponding 1808 VMAT plan

angle-optimized plans achieved dose plans similar to Median follow-up was 6 months, in which time ten
equispaced nine-field plans but with less MUs deliv- (42%) had developed persistent cough requiring
ered (Liu et al. 2006). Further, all IMRT plans had medication and six (25%) developed grade 1–2
reduced V5, V10, V20, and MLD in comparison to ‘‘asymptomatic pneumonia.’’ There were no grade C3
comparable 3D-CRT plans (Liu et al. 2006). adverse reported. Treatment time was 133 ± 7 s
(Scorsetti et al. 2010).
McGrath et al. recently reported on the utility of
3.4 Volumetric-Modulated Arc Therapy VMAT for SBRT. In this study, 21 patients previ-
Experience ously treated with non-coplanar 3D-CRT were
r-planned with a 1808 VMAT hemi-arc (Fig. 8),
Toxicity and outcome data with IMAT and VMAT oriented to avoid contralateral lung. VMAT improved
techniques is notably sparse at the time of this writ- conformity (Table 3; Fig. 9) and decreased lung V5,
ing. Bedford et al. (2008) reported treatment of one V10, V12.5, V20 (Table 1) with equivalent PTV cover-
patient with NSCLC treated to 50 Gy with single-arc age. Although VMAT plans resulted in marginal
VMAT. Compared to a created 3D-CRT plan, the increases in MUs compared to single-segment DMPO
VMAT plan had decreased lung V20 (31.5 vs. 34.8%), plans (5.6%, P = 0.04), the treatment time was
MUs (271 vs. 377), treatment time (90 vs. 180 s), and markedly reduced (6 vs. 12 min, P \ 0.01) (McGrath
improved minimum PTV dose. In a later publication, et al. 2010). A similar planning study from the
Bedford and Warrington (2009) used VMAT to replan Netherlands showed that back-and-forth partial-arc
ten NSCLC patients originally treated to 65 Gy with VMAT plans are comparable to non-coplanar limited-
3D-CRT. Single-arc plans were created spanning segment IMRT for delivery of SBRT but require
3408 at 108 intervals. Results are included in Table 1. significantly less treatment time, 6.5 vs. 23.7 min for
Notably, VMAT plans reduced lung V10, though PTV a single 18 Gy fraction (Holt et al. 2011).
coverage (93 vs. 91%, P = 0.1) and lung V20 were
slightly inferior (Bedford and Warrington 2009).
Scorsetti et al. (2010) reported a series of 24 4 Treatment Planning
consecutive patients with inoperable, stage III
NSCLC treated with 66 Gy using VMAT with two Although the evolution away from elective nodal
partial arcs, avoiding entry through the contralateral irradiation to targeting only gross disease (tumor plus
lung. These plans had excellent PTV coverage (PTV involved lymph nodes) has been slow, it is now the
D99 = 97 ± 2%) and met most planning objectives. accepted standard of care for definitive management
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 701

Table 3 Conformity comparison: 3D-CRT versus IMRT and VMAT


Study Parameters 3D-CRT IMRT VMAT P
Liu et al. (2006) CI 100% 1.5 1.3a – –
McGrath et al. (2010) CI 95% 1.25 – 1.23 NS
McGrath et al. (2010) CI 80% 1.93 – 1.87 0.08
Ong et al. (2010) CI 80% 1.18 – 1.10 0.001
Ong et al. (2010) CI 60% 2.30 – 2.11 0.001
McGrath et al. (2010) CI 50% 5.65 – 5.19 0.01
Ong et al. (2010) CI 40% 4.86 – 5.00 NS
CI conformity index, CI x% = volume of x% isodose/PTV volume, NS not statistically significant
a
Using optimized seven-field IMRT

Fig. 9 Axial isodose comparison: 3D-CRT (left) versus VMAT (right). The VMAT plan (right) reveals improved conformity at
lower isodose lines

in NSCLC. IMRT allows the potential for further of a complete respiratory cycle. CT-PET fusion—
dose-escalation to the target and is now permitted in ideally with the patient in the treatment position for
national cooperative group research protocols. RTOG PET—is used in all cases for tumor and nodal target
Protocol 0617 (ongoing) is the first RTOG protocol volume delineation. As treatment delay can lead to
permitting IMRT delivery. In this two-by-two phase disease progression, if a staging PET has not been
III randomized trial for stage IIIA–B NSCLC, performed within 6–8 weeks of treatment planning,
RT to either 60 Gy (standard arm) versus 74 Gy the PET is generally repeated for re-staging and
(dose-escalated arm) is given concurrently with car- planning purposes (Mohammed et al. 2011a).
boplatin/paclitaxel ± cetuximab followed by further Although respiratory-gating and breath-hold strat-
chemotherapy (Bradley et al. 2007). egies may be preferential to an adaptive image-
guidance strategy in a capable patient, the superiority
likely holds clinical relevance if tumor excursion is
4.1 Simulation approximately 1.4 cm or greater (Hugo et al. 2007a),
quite rare in our experience. Furthermore, 4D-inverse
At our institution, patients are simulated supine in an planning has resulted in plans comparable to real-time
alpha cradle, arms above head using 4D-CT with target tracking methods (Zhang et al. 2008). Gating
3 mm slices from the neck through the liver. Ten CT or breath-hold strategies are not routinely employed
phases are populated to correspond to different phases in our clinic. We do routinely assess for benefit of
702 I. S. Grills and V. S. Mangona

abdominal compression in patients treated with for tumor microscopic extension, a 0.5–0.8 cm
SBRT, though it has rarely been implemented due to (typically 0.5 cm) 3D-CTV margin (excluding bony
lack of observable benefit upon comparing 4D-CT anatomy) is added to GTVITV-primary, creating a
with and without abdominal compression in these CTVITV. Notably, a pathologic study of T1N0 ade-
patients. Thus, we have favored free-breathing treat- nocarcinomas suggests a margin as small as 1.2 mm
ment utilizing an ‘‘internal target volume’’ (ITV) from GTV (on lung windows) to CTV may be suffi-
approach to account for tumor motion and have cient for typical cases, but a 9 mm margin may be
implemented online image-guidance to decrease required to cover 90% of tumors (Grills et al. 2007).
planning treatment volumes. Treatment planning is No CTV margin is added to the nodal GTV
performed on the average phase CT scan, which (CTVnodal = GTVnodal). Similar to RTOG 0617, we
closely corresponds to the mean tumor position at the do not use elective nodal irradiation. For SBRT cases,
time of online cone-beam CT soft tissue registration we use a smaller CTV expansion, 0.3–0.5 cm, given
(Hugo et al. 2007b). the high-dose penumbra in such treatments (Grills
et al. 2007).
The planning target volume (PTV) accounts both
4.2 Target Volumes (Gross Tumor for internal tumor motion as well as setup error.
Volume, Clinical Target Volume, Various methods to account for tumor motion include
Planning Target Volume) (1) an ITV approach, (2) a maximal intensity pro-
jection (MIP) approach, (3) automatic breath-hold, (4)
As per the RTOG 0617 protocol, the gross tumor respiratory gating, and (5) fluoroscopy. RTOG 0617
volume (GTV) includes both primary tumor and recommends fluoroscopy to assess maximal respira-
clinically positive lymph nodes (short axis [1 cm on tory excursion when 4D-CT is not available. With the
CT or PET SUV[3), but no elective nodal irradation. various approaches to account for tumor motion,
If using an ITV approach to account for tumor RTOG 0617 defines minimum CTV-to-PTV expan-
motion, the GTVITV is defined as the volume sions as small as 0.5 cm in all directions—with daily
encompassing the gross tumor throughout a complete bony registration and ITV planning—and up to
respiratory cycle. The clinical target volume (CTV) 1.5 cm (superior–inferior) and 1.0 cm (axial) for
includes an expansion from GTV to account for free-breathing (non-ITV) plans (Table 4).
microscopic extension, defined as a 0.5–1.0 cm
expansion of the entire GTV (or GTVITV) (Bradley
et al. 2007). 4.3 Inverse Planning
At Beaumont, as done on our in-house protocol for
LA-NSCLC, two separate GTV volumes are con- As compared to conventional 3D-CRT, which is
toured on the maximal inspiration phase (phase 0%): forward-planned by choosing a beam angle/weight
GTVprimary (the primary tumor) on lung windows and and aperture size, IMRT is inversely planned by
GTVnodal (inclusive of clinically positive regional providing the treatment planning computer with a
lymph nodes) on mediastinal windows. The GTV is series of objectives and constraints for normal tissues
contoured on a single phase of the respiratory cycle, and target, subsequently allowing the computer to
then propagated to the remaining nine phases fol- generate segment apertures and weights and beam
lowed by physician verification of auto-propagations. weights that provide the ideal fluence map for a given
The union of the GTVprimary from all ten phases plan. Older IMRT planning systems required creation
constitutes the GTVITV-primary. As per our in-house of this ideal fluence map first followed by generation
protocol guidelines, lymph nodes C2.0 cm (long axis) of deliverable segments as a second step. More
on CT or 1.0–1.9 cm (long axis) with increased FDG modern software, however, allows for direct machine
uptake are always included in GTVnodal. Non-FDG- parameter optimization (DMPO), where the genera-
avid, subcentimeter lymph nodes are always exclu- tion of a fluence map and segments are done simul-
ded. Inclusion of FDG-avid subcentimeter lymph taneously considering the predefined constraints of
nodes and non-FDG-avid 1.0–1.9 cm lymph nodes is the treatment machine, substantially shortening the
left to the treating physician’s discretion. To account required time for IMRT planning. Inverse treatment
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 703

Table 4 CTV to PTV expansions: RTOG 0617 (Bradley et al. 2007) and Beaumont methods
RTOG 0617 Beaumont
Free- Breath-hold/ ITV ITV 4D-Adaptive online IGRT
breathing respiratory protocol
gating
No ITV With daily No daily bone With daily With bone or soft (Includes ITV technique
(cm) imaging registration bony tissue registration and bone or soft tissue
(cm) (cm) registration (cm) registration)
(cm)
Superior– C1.5 C1.0 C1.0 C0.5 0.5 Superior: variablea
inferior Motion: Inferior: variablea
C1.0
Setup:
0.5
Axial C1.0 C0.5 C1.0 C0.5 0.5 Anterior: variablea
Posterior: variablea
Motion: Medial: 0.3 cm
C0.5
Setup: Lateral: 0.3 cm
0.5
RTOG radiation therapy oncology group, Motion internal tumor motion, ITV internal target volume, IGRT image-guided radiation
therapy
a
Margin in superior, inferior, anterior, and posterior directions determined according to the adaptive process

planning software takes the prescribed dose distribu-


tion and optimizes a plan by minimizing the ‘‘cost 5 Targeting and Verification
function,’’ a function that quantifies variance from the
pre-determined dose–volume histogram (DVH) 5.1 Image-Guided Radiation Therapy
objectives.
Dose constraints from RTOG 0617 (Bradley et al. In an effort to accurately target disease, escalate dose,
2007) and RTOG 0839 (Edelman et al. 2010) are and spare organs at risk, two main targeting strategies
seen in Table 5. Although planning the best treat- are routinely implemented: biological targeting with
ment plan for a particular case inevitably involves positron emission tomography (PET) and image-
manual iteration, Craft et al. (2007) developed a guidance. New 4D-imaging and planning techniques
technique that generates a database of optimal plans further facilitate improvements in radiotherapy
from which the user may select the plan felt clini- administration. Harsolia et al. compared conventional
cally preferential. 3D-CRT plans—using free-breathing planning CT
For arc therapy planning (either IMAT or and fluoroscopy to assess tumor motion—to new 4D-
VMAT), MLC positions must be contiguous, and techniques using combinations of 4D-imaging, 4D-
incremental control points (e.g. every 6) are used planning, and image-guided radiation therapy (IGRT)
from which interval data can be interpolated. with oonboard 4D-conebeam CT (4D-CBCT) using
Multiple inverse planning techniques have now been three strategies: (1) 4D-union technique, (2) 4D off-
developed for both single- and multi-arc treatments, line adaptive planning with a single correction (offline
facilitating VMAT planning and delivery, thereby adaptive radiotherapy (ART)), and (3) 4D-online
allowing this new modality to become adapted into adaptive planning with daily correction (online ART)
clinical practice (Otto 2008; Luan et al. 2008; Wang (Harsolia et al. 2008). The 4D-union plan used an ITV
et al. 2008; Cao et al. 2009). At Beaumont, VMAT technique, defining GTVITV as the union of the GTV
plans are generated with SmartArc, integrated in phases with 5 mm expansions for CTV and PTV. The
Pinnacle3 version 9. ART process defines a probability density function
704 I. S. Grills and V. S. Mangona

Table 5 Dose constraints to organs at risk


RTOG 0617 (Bradley et al. 2007) RTOG 0839 (Edelman et al. Beaumont Beaumont
(60 vs. 74 Gy) 2010) (60 Gy) (2.0 Gy/day) (1.5 Gy BID)
Spinal cord Dmax \ 50.5 Gya Dmax \ 50.5 Gya Dmax(+3 mm) B Dmax(+3 mm) B 47
50 Gya Gya
Lungs (Excludes CTV) (Excludes CTV) (Excludes GTV) (Excludes GTV)
V20 \ 37% (or) V20 \ 37% (or) V20 B 30% (and) V18.7 B 30%
(and)
Dmean \ 20 Gy Dmean \ 20 Gy Dmean B 18 Gy Dmean B 17 Gy
b
Esophagus Dmean \ 34 Gy Dmax \ 40 Gy V50 B 30% V47 B 30%
Dmean B 25 Gy Dmean B 23.4 Gy
Dmax(+3 mm) B Dmax(+3 mm) B
75 Gy 70 Gy
Brachial Dmax \ 66 Gy Dmax \ 66 Gy Dmax B 66 Gy Dmax B 63 Gy
plexus
Heart D33 \ 60 Gyc D33 \ 60 Gy V50 B 50% V47 B 50%
D66 \ 45 Gyc D66 \ 45 Gy D33 B 66 Gy D33 B 62 Gy
D100 \ 40 Gyc D100 \ 40 Gy D100 B 40 Gy D100 B 37.5 Gy
BID twice daily fractions, Dmax maximum dose, Dmean mean dose, +3 mm = volume ? 3 mm margin, Vx proportional volume of
the region of interest receiving Cx Gy, Dy dose delivered to y% of the region of interest
a
Required constraint. For RTOG 0839, maximum dose is defined as maximum dose for contiguous volume C0.03 ml
b
constraint is strongly recommended
c
constraint recommended but not required

Table 6 Mean relative reduction compared to standard 3D-CRT plan (adapted from Harsolia et al. 2008)
Parameter 3D- 4D-union 4D-offline ART (single correction) 4D-online ART (daily correction)
CRT (%) (%) (%)
PTV volume – ;15 ;39 ;44
Lungs V20 (%) – ;21 ;23 ;31
Mean lung dose – ;16 ;26 ;31
3D-CRT 3D-conformal radiation therapy, ART adaptive radiation therapy, PTV planning target volume, online ART adaptive with
daily correction, offline ART adaptive plan with a single correction, V20 (%) = % volume of lungs excluding GTV receiving
C20 Gy

(PDF) of tumor position vs. time estimated by fluo- significant with daily online adaptive IGRT (Table 6;
roscopy and/or 4D-CT or 4D-CBCT to account for Harsolia et al. 2008).
tumor motion and interfraction variability of tumor
location. The offline ART process uses a single
adjustment after the first week of treatment, 5.2 Biological Targeting with Positron
accounting for individualized variability assessed Emission Tomography
after five fractions. The online ART process used a
daily fluoroscopy-guided correction to account for Since FDA approval of 18-fluorodeoxyglucose posi-
daily setup error, thus the PDF still accounted for tron emission tomography (18FDG-PET) for staging
respiratory motion, but accounted for less setup error lung cancer, its utility has continued to expand
than with the offline technique, further decreasing (Coleman and Tesar 1997). PET facilitates biological
the CTV-to-PTV expansion. IMRT plans decreased targeting and is now used in all phases of radiation
PTV volume, mean lung dose, and lung V20, most therapy from staging to planning to follow-up. PET
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 705

has proven its role in NSCLC staging, with greater phantom. Ion chambers and films are used to verify
sensitivity and accuracy than CT (Dwamena et al. the dose and spatial dose gradient, respectively.
1999). PET helps define clinically positive lymph Alternatively, individual beams can be assessed with
node regions and define tumor extent, complementary two-dimensional (2D) arrays of diodes such as the
to the anatomic information provided by CT imaging MapcheckTM (Sun Nuclear). Electronic portal imag-
alone. Studies have shown that PET-to-CT (simula- ing devices (EPIDs) serve to create beam’s-eye-view
tion/planning CT) registration can result in smaller portal images for user verification of beam position
target volumes secondary to improved GTV definition and treatment setup. More recently, EPID images
but also upstaging, leading to increased target volume have been shown to efficiently carry out the absolute
(Giraud et al. 2001; Bradley et al. 2004; Erdi et al. dosimetry for IMRT quality assurance with the added
2002; Mah et al. 2002; Ung et al. 2000). With such benefit of improved spatial resolution lacking in
biological targeting afforded by PET, the risk of target 2D-diode arrays (Nelms et al. 2010). 3D-diode arrays
miss can be decreased while facilitating dose-escala- such as the Delta4 phantom have now come into
tion by shrinking treatment volume, two key goals of utility for quality assurance with IMRT and VMAT
IMRT. As PET-CT scanners have become more treatments, obtaining accurate 3D-absolute dosimetry
routinely available, patients have CT-simulation without ionization chambers or film (Bedford and
immobilized in the treatment position and then pro- Young 2009). Online volumetric (3D) pre-treatment
ceed to the planning PET-CT. For planning, the imaging (e.g. with online CBCT registration to the
simulation acts as the primary data set to which the planning CT) serves as an ideal method for target
CT portion of the PET-CT is merged, usually with verification at the time of treatment. This is particu-
bony registration. PET has further been utilized to larly important in the case of IMRT or SBRT where
assess tumor response to radiation therapy (Moham- the dose falloff between target and normal tissues is
med et al. 2011b) and potentially predict local failure sharp and the total number of treatment fractions may
(Mangona et al. unpublished abstract). Follow-up be limited.
PET-CT data from Wong et al. revealed significant
reduction in maximum SUV (standardized uptake
value) and CT size after treatment, with relative 6 Outcomes
reductions in maximal metabolic uptake greater than
size reduction (62 vs. 47%, P = 0.03) (Wong et al. With the routine implementation of IMRT being rel-
2007). atively recent, data from prospective clinical studies
Recently, our SBRT and LA-NSCLC protocols treated with this modality are still maturing (RTOG
have implemented weekly on-treatment respiratory- 0617 is still open to accrual at the time of this writ-
gated 4D-PET-CT. 4D-PET-CTs are performed ing). However, IMRT has shown ability to escalate
including gated and non-gated sequences 60 and tumor dose while sparing normal tissue. In the ret-
120 min after injection (early and delayed scans) rospective study from MD Anderson including
including both TOF (time-of-flight) and non-TOF patients with LA-NSCLC treated with concurrent
processing. By assessing early RT-tumor metabolic chemoradiotherapy (mean dose 63 Gy), Yom et al.
response, on-treatment PET may potentially identify (2007) analyzed outcomes of the subgroup receiving
patients who could ultimately benefit from some form IMRT (n = 68). For 6- and 12-month follow-up,
of additional treatment. locoregional control (LRC) was 94 and 55%, disease-
free survival (DFS) 67 and 32%, and overall survival
(OS) 79 and 57%, respectively. The median follow-up
5.3 Treatment Verification was only 8 months.
Sura et al. (2008) similarly reported the MSKCC
With increased conformity, decreased margins, and experience, including 55 patients with inoperable
intensity-modulation comes increased dependence on stage I–IIIB disease treated with IMRT. The median
quality assurance. IMRT and VMAT plans require follow-up in this study was significantly longer,
dosimetric verification prior to delivery (Ezzell et al. 21 months. Two-year local control (LC) rates were
2003). Traditionally, the plan can be delivered to a 50% (stage I–II, n = 16) and 58% (stage III, n = 29).
Table 7 Outcomes in NSCLC treated with IMRT and VMAT
706

Study n Time Patients Dose (Gy) Gpf RT Chemotherapy Follow-up Parameter Outcome
mean/median (Gy) Type median
(range) (range)
MSKCC (Sura 55 2001–2005 Inoperable 69.5 (60–90) 1.8–2.0 IMRT Sequential 21 months Local Control 2 year: 50%
et al. 2008) Stage (53%) (0–61) (Stage I–II)
I–IIIB Concurrent 2 year: 58%
(24%) (Stage III)
MD Anderson 68 2002–2005 Stage 63 (50.4–76) 1.8–2.0 IMRT Induction 8 months Locoregional Control 6 months:
(Yom et al. 2007) IIB–IV (29%) (0–27) 94%
Concurrent 12 months:
(100%) 55%
Belgium (Bral 40 2005–2008 Inoperable 70.5 (all) 2.35 Helical Sequential 14 monthsa LPFS 1 year: 66%
et al. 2010) Stage IIIa TT (82%)
Concurrent 2 year: 50%
(0%)
None (18%)
Milan (Scorsetti 24 2009 Inoperable 66 (50–66) 2.0 VMAT Concurrent 6 months Response at 3 months PR [ 50%:
et al. 2010) Stage III (46%) (RECIST(Therasse et al. 56%
Sequential 2000)) PR \ 50%:
(33%) 22%
None (21%) Stable
disease: 22%
Disease
progression:
0%
Beaumont 17 2008–2010 Stage III 66 (66–72, 54 1.5 IMRT Concurrent 12 months LR 1 year: 6%
(abstract) (McGee preoperative) BID (100%)
et al. 2010)
Beaumont 17 2008–2010 Stage III 66 (66–72, 54 1.5 IMRT Concurrent 12 months RR 1 year: 30%
(abstract) (McGee preoperative) BID (100%)
et al. 2010)
Beaumont 17 2008–2010 Stage III 66 (66–72, 54 1.5 IMRT Concurrent 12 months DM 1 year: 31%
(abstract) (McGee preoperative) BID (100%)
et al. 2010)
(continued)
I. S. Grills and V. S. Mangona
Table 7 (continued)
Study n Time Patients Dose (Gy) Gpf RT Chemotherapy Follow-up Parameter Outcome
mean/median (Gy) Type median
(range) (range)
Belgium (Bral 40 2005-2008 Inoperable 70.5 (all) 2.35 Helical Sequential 14 monthsa DMFS Median:
et al. 2010) Stage III TT (82%) 10.6 months
Concurrent
(0%)
None (18%) 1 year: 43%
MD Anderson 68 2002-2005 Stage 63 (50.4–76) 1.8–2.0 IMRT Concurrent 8 months DFS 6 months:
(Yom et al. 2007) IIB-IV (100%) (0–27) 67%
12 months:
32%
MSKCC (Sura 55 2001–2005 Inoperable 69.5 (60–90) 1.8–2.0 IMRT Sequential 21 months CSS 2 year: 63%
et al. 2008) Stage (53%) (0–61)
I–IIIB Concurrent
(24%)
MSKCC (Sura 55 2001–2005 Inoperable 69.5 (60–90) 1.8–2.0 IMRT Sequential 21 months OS Median:
et al. 2008) Stage (53%) (0–61) 25 months
I–IIIB 2 year: 57%
Concurrent 2 year: 55%
(24%) (Stage I–II)
2 year: 58%
(Stage III)
MD Anderson 68 2002–2005 Stage 63 (50.4–76) 1.8–2.0 IMRT Induction 8 months OS 6 months:
(Yom et al. 2007) IIB–IV (29%) (0–27) 79%
Concurrent 12 months:
(100%) 55%
(continued)
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer
707
Table 7 (continued)
708

Study n Time Patients Dose (Gy) Gpf RT Chemotherapy Follow-up Parameter Outcome
mean/median (Gy) Type median
(range) (range)
Belgium (Bral 40 2005–2008 Inoperable 70.5 (all) 2.35 Helical Sequential 14 monthsa OS Median:
et al. 2010) Stage III TT (82% 17 months
Concurrent Median:
(0%) 21 months
(IIIA)
None (18%) Median:
12 months
(IIIB)
1 year: 65%
2 year: 27%
Beaumont 17 2008–2010 Stage III 66 (66–72, 54 1.5 IMRT Concurrent 12 months OS 1 year: 92%
(abstract) (McGee preoperative) BID (100%)
et al. 2010)
NSCLC non-small-cell lung cancer, IMRT intensity-modulated radiation therapy, VMAT volumetric-modulated radiation therapy, MSKCC Memorial Sloan-Kettering Cancer
Center, Gpf gray per fraction, BID twice daily, LPFS local progression-free survival, LR local recurrence, PR partial remission, RR regional recurrence, DFS disease-free survival,
DMFS distant-metastasis-free survival, CSS cause-specific survival, OS overall survival, TT tomotherapy
a
Median follow-up = 14 months in 16 survivors
I. S. Grills and V. S. Mangona
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 709

For all patients, median disease-free survival (DFS)


was 12 months, and 2-year DFS was 41%. Two-year 7 Conclusion
cancer-specific survival (CSS) and OS were 63 and
57%, respectively. Survival was similar in stage I–II Over the past ten years, technological advancements
and stage III patients with a median of 25 months. have brought IMRT into routine clinical use with
The largest published prospective trial of NSCLC inclusion in lung RTOG protocols since 2007. In this
patients treated with IMRT is a Belgian trial of hyp- technically challenging and prognostically poor dis-
ofractionated RT using helical tomotherapy (70.5 Gy, ease, dose-escalation and normal tissue sparing have
2.35 Gy per fraction) in 40 consecutive patients with been shown to be imperative for safely improving local
inoperable IIIA (n = 16) and IIIB (n = 24) NSCLC disease control while minimizing risk for potentially
from 2005 to 2008. Patients were permitted induction severe acute and chronic toxicity. From the planning
chemotherapy, but not concurrent with RT. Overall, perspective, IMRT has long proven its superiority to
1- and 2-year local progression-free survival was 66 3D-CRT for safe dose-escalation in properly selected
and 50%, respectively. Median time to distant cases. As advancements such as biological targeting
metastasis was 10.6 months, and 1-year distant- improve our ability to define clinical targets and
metastasis-free survival (DMFS) was 43%. Median advancements such as online image-guidance to reduce
OS was 21 versus 12 months for IIIA and IIIB disease target miss and PTV volumes, so may the full clinical
(P = 0.03) and 17 months for all patients. Two potential of IMRT—particularly in the setting of
patients (5%) died of acute pulmonary toxicity; LA-NSCLC—be better attained. Although still early in
median follow-up in 14 survivors was 16 months clinical use, VMAT offers a new, promising way to face
(Bral et al. 2010). the challenges set forth by lung cancer. With inverse-
In Scorsetti’s VMAT study (limited follow-up) of planning algorithms and quality-assurance methods
24 patients with large-volume, unresectable, stage III available, its utility has grown rapidly in the past few
NSCLC, patients received 66 Gy delivered with two years. Initial studies have shown the potential for high
isocentric partial arcs. Using the RECIST (2000) quality treatment plans with significantly curtailed
(Therasse et al. 2000) criteria for assessing response treatment time. Although lung cancer radiotherapy has
to solid tumors, at 3-month follow-up, 14 (56%) had substantially improved in the past ten years, outcomes
partial remission [50%, five (22%) had partial with LA-NSCLC are still poor. Although the dosi-
remission \50%, five (22%) had stable disease, and metric advantages of IMRT are quite clear, clinical
no patients had evidence of disease progression outcomes of patients treated with IMRT continue to
(Scorsetti et al. 2010). mature with prospective data only recently appearing
At Beaumont, 17 patients from 2008 to 2010 were as full articles in peer-reviewed journals. We have
enrolled to a prospective, hyperfractionated, dose- optimism that technological advances (e.g. IMRT,
escalation trial for patients with stage IIIA–IIIB VMAT, 4D-image-guidance/planning) coupled with
NSCLC using online 4D-adaptive CBCT image- future advances will translate into significant clinical
guidance. Non-surgical patients received 66–72 Gy, benefit for what continues to be the most common cause
preoperative patients 54 Gy. Fractions were 1.5 Gy of cancer-related death in the US and worldwide.
twice daily with a 6 h interfraction interval using
IMRT. With median follow-up of 12 months, pre-
liminary outcomes appear quite favorable with 1-year
incidences of local recurrence, regional recurrence, References
and distant metastasis of 6, 30, and 31%, respectively.
A single patient who had developed distant metastasis American Cancer Society: Cancer Facts And Figures (2010)
died of disease. With the remaining 16 patients cur- American Cancer Society, Atlanta, Ga. http://www.cancer.
rently still living, 1-year overall survival is 92%. org/acs/groups/content/@nho/documents/document/acspc-
(McGee et al. 2010). A summary table of outcomes 024113.pdf. Accessed 19 Feb 2011
Armstrong JG, Minsky BD (1989) Radiation therapy for
for patients with LA-NSCLC treated with IMRT is medically inoperable stage I, II non-small-cell lung cancer.
seen in Table 7. Cancer Treat Rev 16(4):247–255
710 I. S. Grills and V. S. Mangona

Arriagada R, Le Chevalier T, Quoix E, Ruffie P, de Cremoux H, arc therapy. Phys Med Biol 54:6725–6738. doi:10.1088/
Douillard JY, Tarayre M, Pignon JP, Laplanche A (1991) 0031-9155/54/21/018
ASTRO (American Society for Therapeutic Radiology and Chapet O, Thomas E, Kessler ML, Fraass BA, Ten Haken RK
Oncology) plenary: effect of chemotherapy on locally (2005) Esophagus sparing in lung tumor irradiation: an
advanced non-small-cell lung carcinoma: a randomized EUD-based optimization technique. Int J Radiat Oncol Biol
study of 353 patients. GETCB (Groupe d’Etude et Traite- Phys 63:179–187. doi:10.1016/j.ijrobp.2005.01.028
ment des Cancers Bronchiques), FNCLCC (Féderation Chapet O, Fraass BA, Ten Haken RK (2006) Multiple fields
Nationale des Centres de Lutte contre le Cancer) and the may offer better esophagus sparing without increased
CEBI trialists. Int J Radiat Oncol Biol Phys 20:1183–1190 probability of lung toxicity in optimized IMRT of lung
Ataman ÖU, Bentzen SM, Saunders MI, Dische S (2001) Cancer tumors. Int J Radiat Oncol Biol Phys 65:255–265. doi:
research campaign failure-specific prognostic factors after 10.1016/j.ijrobp.2005.12.028
continuous hyperfractionated accelerated radiotherapy Choy H, Akerley W, Safran H, Graziano S, Chung C, Williams
(CHART) or conventional radiotherapy in locally advanced T, Cole B, Kennedy T (1998) Multiinstitutional phase II
non-small-cell lung cancer: a competing risks analysis. Br J trial of paclitaxel carboplatin and concurrent radiation
Cancer 85(8):1113–1118. doi:10.1054/bjoc.2001.2049 therapy for locally advanced non-small-cell lung cancer.
Bedford JL, Warrington AP (2009) Commissioning of volu- J Clin Oncol 16:3316–3322
metric modulated arc therapy (VMAT). Int J Radiat Oncol Clark GM, Popple RA, Young PE, Fiveash JB (2010)
Biol Phys 73:537–545. doi:10.1016/j.ijrobp.2008.08.055 Feasibility of single-isocenter volumetric modulated arc
Bedford JL, Young K (2009) Evaluation of the delta4 phantom for radiosurgery for treatment of multiple brain metastases. Int J
IMRT and VMAT verification. Phys Med Biol 54:N167– Radiat Oncol Biol Phys 76:296–302. doi:10.1016/j.ijrobp.
N176. doi:10.1088/0031-9155/54/9/N04 2009.05.029
Bedford J, Nordmark Hansenv V, McNair H, Aitken AH, Brock Coleman RE, Tesar RD (1997) Clinical PET: Are we ready?
JEC, Warrington AP, Brada M (2008) Treatment of lung J Nucl Med 38:16N–24N
cancer using volumetric modulated arc therapy and image- Cox JD, Stetz J, Pajak TF (1995) Toxicity criteria of the
guidance: a case study. Acta Oncol 47:1438–1443. doi: radiation therapy oncology group (RTOG) and the European
10.1080/02841860802282778 Organization For Research And Treatment Of Cancer
Bortfield T (2006) IMRT: a review and preview. Phys Med Biol (EORTC). Int J Radiat Oncol Biol Phys 31:1341–1346. doi:
51:R363–R379. doi:10.1088/0031-9155/51/13/R21 10.1016/0360-3016(95)00060-C
Bradley J et al (2004) Impact of FDG-PET on radiation therapy Craft D, Halabi T, Shih HA, Bortfeld T (2007) An approach
volume delineation in non-small-cell lung cancer. Int J for practical multiobjective IMRT treatment planning. Int J
Radiat Oncol Biol Phys 59:78–86 Radiat Oncol Biol Phys 69:1600–1607. doi:10.1016/j.ijrobp.
Bradley et al (2007) RTOG 0617 a randomized phase III 2007.08.019
comparison of standard-dose (60 Gy) versus high dose Curran W, Scott C, Langer C, Komaki R, Lee JS, Hauser S,
(74 Gy) conformal radiotherapy with concurrent and consol- Movsas B, Wasserman T, Sause W, Cox JD (2003) Long-
idation carboplatin/paclitaxel ± cetuximab (IND #103444) in term benefit is observed in a phase III comparison of
patients with stage IIIA/IIIB non-small-cell lung cancer. sequential versus concurrent chemo-radiation for patients
http://www.rtog.org/members/protocols/0617/0617.pdf. with unresectable NSCLC: RTOG 9410 [Abstract 2499].
Accessed 4 Feb 2011 Proc Am Soc Clin Oncol 22:621a
Bradley J, Graham MV, Winter K, Purdy JA, Komaki R, Roa Dosoretz DE, Katin MJ, Blitzer PH et al (1996) Medically
WH, Ryu JK, Bosch W, Emami B (2005) Toxicity and inoperable lung carcinoma: the role of radiation therapy.
outcome results of RTOG 9311: a phase I–II dose-escalation Semin Radiat Oncol 6(2):98–104
study using three-dimensional conformal radiotherapy in Dwamena BA, Sonnad SS, Angobaldo JO, Wahl RL (1999)
patients with inoperable non-small-cell lung carcinoma. Int Metastases from non-small-cell lung cancer: mediastinal
J Radiat Oncol Biol Phys 61:318–328. doi:10.1016/j.ijrobp. staging in the 1990s—meta-analytic comparison of PET and
2004.06.260 CT. Radiology 213(2):530–536
Brahme A (1988) Optimization of stationary and moving beam Edelman et al (2010) RTOG 0839 randomized phase II study of
radiation therapy. Radiother Oncol 12:129–140 pre-operative chemoradiotherapy ± panitumumab (IND
Bral S, Duchateau M, Versmessen H, Engels B, Tournel K, #110152) followed by consolidation chemotherapy in
Vinh-Hung V, De Ridder M, Schallier D, Storme G (2010) potentially operable locally-advanced (stage IIIA, N2+),
Toxicity and outcome results of a class solution with non-small-cell lung cancer. http://www.rtog.org/members/
moderately hypofractionated radiotherapy in inoperable protocols/0839/0839.pdf. Accessed 19 Feb 2011
stage III non-small cell lung cancer using helical tomother- Erdi YE et al (2002) Radiotherapy treatment planning for
apy. Int J Radiat Oncol Biol Phys 77:1352–1359. doi: patients with non-small-cell lung cancer using positron
10.1016/j.ijrobp.2009.06.075 emission tomography (PET). Radiother Oncol 62:51–60
Cao D, Holmes TW, Afghan M, Shepard DM (2007) Compar- Ezzell GA, Galvin JM, Low D, Palta JR, Rosen I, Sharpe MB, Xia
ison of plan quality provided by intensity-modulated arc P, Xiao Y, Xing L, Yu CX (2003) Guidance document on
therapy and helical tomotherapy. Int J Radiat Oncol Biol delivery, treatment planning, and clinical implementation of
Phys 69:240–250. doi:10.1016/j.ijrobp.2007.04.073 IMRT: report of the IMRT subcommittee of the AAPM
Cao D, Afghan MK, Ye J, Chen F, Shepard DM (2009) A radiation therapy committee. Med Phys 30:2089–2115. doi:
generalized inverse planning tool for volumetric-modulated 10.1118/1.1591194
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 711

Fay M, Tan A, Fisher R, Mac Manus M, Wirth A, Ball D stereotactic body radiotherapy of lung tumors: a comparison
(2005) Dose–volume histogram analysis as predictor of with intensity-modulated radiotherapy techniques. Int J
radiation pneumonitis in primary lung cancer patients Radiat Oncol Biol Phys (ePub 2011 Feb 5). doi:10.1016/
treated with radiotherapy. Int J Radiat Oncol Biol Phys j.ijrobp.2010.09.014
66:1355–1363. doi:10.1016/j.ijrobp.2004.08.025 Hugo GD, Yan D, Liang J (2007a) Population and patient-specific
Furuse K, Fukuoka M, Kawahara M, Nishikawa H, Takada Y, target margins for 4D adaptive radiotherapy to account for
Kudoh S, Katagami N, Ariyoshi Y (1999) Phase III study intra- and inter-fraction variation in lung tumour position.
of concurrent versus sequential thoracic radiotherapy in Phys Med Biol 52:257. doi:10.1088/0031-9155/52/1/017
combination with mitomycin, vindesine, and cisplatin in Hugo GD, Liang J, Campbell J, Yan D (2007b) On-line target
unresectable stage III non-small-cell lung cancer. J Clin position localization in the presence of respiration: a
Oncol 17:2692–2699 comparison of two methods. Int J Radiat Oncol Biol Phys
Giraud P et al (2001) CT and (18) F-deoxyglucose (FDG) 69:1634-1641. doi:10.1016/j.ijrobp.2007.08.023
image fusion for optimization of conformal radiotherapy of Kaskowitz L, Graham MV, Emami B, Halverson KJ, Rush C
lung cancers. Int J Radiat Oncol Biol Phys 49:1249–1257 (1993) Radiation therapy alone for stage I non-small-cell
Gopal R, Starkschall G, Tucker SL, Cox JD, Liao Z, Hanus M, lung cancer. Int J Radiat Oncol Biol Phys 7(3):517–523
Kelly JF, Stevens CW, Komaki R (2003) Effects of Lau D, Leigh B, Gandara D, Edelman M, Morgan R, Israel V,
radiotherapy and chemotherapy on lung function in patients Lara P, Wilder R, Ryu J, Doroshow J (2001) Twice weekly
with non-small-cell lung cancer. Int J Radiat Oncol Biol paclitaxel and weekly carboplatin with concurrent thoracic
Phys 56:114–120. doi:10.1016/S0360-3016(03)00077-4 irradiation followed by carboplatin/paclitaxel consolidation
Grills IS, Yan D, Martinez AA, Vicini FA, Wong JW, Kestin for stage III non-small-cell lung cancer. a California Cancer
LL (2003) Potential for reduced toxicity and dose-escalation Consortium phase II trial. J Clin Oncol 19:442–447
in the treatment of inoperable non-small-cell lung cancer: Lievens Y, De Wever W, Stroobants S, Van den Heuvel F
a comparison of intensity-modulated radiation therapy (2011) Intensity-modulated radiotherapy for locally
(IMRT), 3D-conformal radiation, and elective nodal irradi- advanced non-small-cell lung cancer: a dose-escalation
ation. Int J Radiat Oncol Biol Phys 57:875–890. doi: planning study. Int J Radiat Oncol Biol Phys 80:306–313.
10.1016/S0360-3016(03)00743-0 doi:10.1016/j.ijrobp.2010.06.025
Grills IS, Fitch DL, Goldstein NS, Yan D, Chmielewski GW, Liu HH, Wang X, Dong L et al (2004) Feasibility of sparing
Welsh RJ, Kestin LL (2007) Clinicopathologic analysis of lung and other thoracic structures with intensity-modulated
microscopic extension in lung adenocarcinoma: defining radiotherapy for non-small-cell lung cancer. Int J Radiat
clinical target volume for radiotherapy. Int J Radiat Oncol Oncol Biol Phys 58:1268–1279
Biol Phys 69:334–341. doi:10.1016/j.ijrobp.2007.03.023 Liu HH, Jauregui M, Zhang X, Wang X, Dong L, Mohan R (2006)
Grills IS, Hope AJ, Guckenberger M, Kestin LL, Werner- Beam angle optimization and reduction for intensity-modu-
Wasik M, Yan D, Sonke J, Bissonnette J, Xiao Y, Belderbos lated radiation therapy of non-small-cell lung cancers. Int J
J (2010a) A Collaborative Analysis of Stereotactic Lun- Radiat Oncol Biol Phys 65:561–572. doi:10.1016/j.ijrobp.
gRadiotherapy (Lung SBRT) Outcomes for Stage I Non- 2006.01.033
small Cell Lung Cancer (NSCLC) using Daily Online Cone- Luan S, Wang C, Cao D, Chen DZ, Shepard DM, Yu CX
beam CT Image-guided Radiotherapy (CBCT-IGRT) (2008) Leaf-sequencing for intensity-modulated arc therapy
[Abstract 30] Int J Radiat Oncol Biol Phys 78:S14. doi: using graph algorithms. Med Phys 35:61–69. doi:10.1118/
10.1016/j.ijrobp.2010.07.075 1.2818731
Grills IS, Mangona VS, Welsh R, Chmielewski G, McInerney Mah K et al (2002) The impact of (18)FDG-PET on target and
E, Martin S, Wloch J, Ye H, Kestin LL (2010b) Outcomes critical organs in CT-based treatment planning of patients
after stereotactic lung radiotherapy or wedge resection for with poorly defined non-small-cell lung carcinoma: a
stage I non-small-cell lung cancer. J Clin Oncol 28:928-935. prospective study. Int J Radiat Oncol Biol Phys 52:339–350
doi: 10.1200/JCO.2009.25.0928 Matuszak MM, Yan D, Grills IS, Martinez A (2010) Potential
Guckenberger M, Richter A, Krieger T, Wilbert J, Baier K, impact of volumetric modulated arc therapy on the planning
Flentje M (2009) Is a single arc sufficient in volumetric- and delivery of radiation therapy. Int J Radiat Oncol Biol
modulated arc therapy (VMAT) for complex-shaped target Phys 77:608–616. doi:10.1016/j.ijrobp.2009.08.032
volumes? Radiother Oncol 93:259–265. doi:10.1016/ Mayo CS, Urie MM, Fitzgerald TJ, Ding L, Lo YC, Bogdanov
j.radonc.2009.08.015 M (2008) Hybrid IMRT for treatment of cancers of the lung
Harsolia A, Hugo GD, Kestin LL, Grills IS, Yan D (2008) and esophagus. Int J Radiat Oncol Biol Phys 71:1408–1418.
Dosimetric advantages of four-dimensional adaptive image- doi:10.1016/j.ijrobp.2007.12.008
guided radiotherapy for lung tumors using online cone- McDermott PN, Orton CG (2010) The physics and technology
beam computed tomography. Int J Radiat Oncol Biol Phys of radiation therapy. Madison, Wisconsin
70:582–589. doi:10.1016/j.ijrobp.2007.08.078 McGee MC, Grills IS, Ionascu D, Wloch J, Martin S, Margolis
Hayman JA, Martel MK, Ten Haken RK, Normolle DP, Todd J, Welsh R, Chmielewski G, Yan D, Kestin LL (2010)
RF III, Littles JF, Sullivan MA, Possert PW, Turrisi AT, Feasibility and toxicity of dose-escalated 4D-adaptive
Lichter AS (2001) Dose-escalation in non-small-cell lung image-guided radiotherapy (IGRT) for non-small-cell lung
cancer using three-dimensional conformal radiation ther- cancer (NSCLC) [Abstract 2706]. Int J Radiat Oncol Biol
apy: update of a phase I trial. J Clin Oncol 19:127–136 Phys 78:S520. doi:10.1016/j.ijrobp.2010.07.1214
Holt A, van Vliet-Vroegindeweij C, Mans A, Belderbos JS, McGrath SD, Matuszak MM, Yan D, Kestin LL, Martinez AA,
Damen EMF (2011) Volumetric-modulated arc therapy for Grills IS (2010) Volumetric modulated arc therapy for
712 I. S. Grills and V. S. Mangona

delivery of hypofractionated stereotactic lung radiotherapy: Rosenzweig KE, Fox JL, Yorke E, Amols H, Jackson A, Rusch V,
a dosimetric and treatment efficiency analysis. Radiother Kris MG, Ling CC, Leibel SA (2005) Results of a phase I dose-
Oncol 95:153–157. doi:10.1016/j.radonc.2009.12.039 escalation study using three-dimensional conformal radiother-
Mohammed N, Kestin LL, Grills IS, Battu M, Fitch DL, Wong apy in the treatment of inoperable non-small-cell lung
CY, Margolis JH, Chmielewski GW, Welsh RJ (2011a) carcinoma. Cancer 103:2118–2127. doi:10.1002/cncr.21007
Rapid disease progression with delay in treatment of non- Schallenkamp JM, Miller RC, Brinkmann DH, Foote T, Garces
small-cell lung cancer. Int J Radiat Oncol Biol Phys YI (2007) Incidence of radiation pneumonitis after thoracic
79(2):466-72. doi: 10.1016/j.ijrobp.2009.11.029 irradiation: dose–volume correlates. Int J Radiat Oncol Biol
Mohammed N, Grills IS, Wong CY, Galerani AP, Chao K, Welsh Phys 67:410–416. doi:10.1016/j.ijrobp.2006.09.030
R, Chmielewski G, Yan D, Kestin LL (2011b) Radiographic Schwarz M, Alber M, Lebesque JV, Mijnheer BJ, Damen EMF
and metabolic response rates following image-guided stereo- (2005) Dose heterogeneity in the target volume and intensity-
tactic radiotherapy for lung tumors. Radiother Oncol 99: modulated radiotherapy to escalate the dose in the treatment
18-22. doi: 10.1016/j.radonc.2011.03.003 of non-small-cell lung cancer. Int J Radiat Oncol Biol Phys
Murshed H, Liu HH, Liao Z, Barker JL, Wang X, Tucker SL, 62:561–570. doi:10.1016/j.ijrobp.2005.02.011
Chandra A, Guerrero T, Stevens C, Chang JY, Jeter M, Cox Scorsetti M, Navarria P, Mancoso P, Alongi F, Castiglioni S,
JD, Komaki R, Mohan R (2004) Dose and volume reduction Cavina R, Cozzi L, Fogliata A, Pentimalli S, Tozzi A,
for normal lung using intensity modulated radiotherapy for Santoro A (2010) Large-volume unresectable locally-
advanced stage non-small-cell lung cancer. Int J Radiat Oncol advanced non-small-cell lung cancer: acute toxicity and
Biol Phys 58:1258–1267. doi:10.1016/j.ijrobp.2003.09.086 initial outcome results with rapid arc. Radiat Oncol 5:94.
Narayan S, Henning GT, Ten Haken RK, Sullivan MA, Martel doi:10.1186/1748-717X-5-94
MK, Hayman JA (2004) Results following treatment to Shaitelman SF, Grils IS, Liang J, Zhuang L, Mangona V, Yan D,
doses of 92.4 or 102.9 Gy on a phase I dose-escalation study Kestin LL (2009) A comprehensive dose–volume analysis of
for non-small-cell lung cancer. Lung Cancer 44:79–88. doi: predictors of pneumonitis and esophagitis following radio-
10.1016/j.lungcan.2003.09.015 therapy for non-small-cell lung cancer (NSCLC) [Abstract
Nelms BE, Rasmussen KH, Tomé WA (2010) Evaluation of a 2636]. Int J Radiat Oncol Biol Phys 75:S468. doi:10.1016/
fast method of EPID-based dosimetry for intensity-modu- j.ijrobp.2009.07.1068
lated radiation therapy. J Appl Clin Med Phys 11(2):3185 Shi A, Zhu G, Wu H, Yu R, Li F, Xu B (2010) Analysis of
Ong CL, Verbakel WF, Cuijpers JP, Slotman BJ, Lagerwaard clinical and dosimetric factors associated with severe acute
FJ, Senan S (2010) Stereotactic radiotherapy for peripheral radiation pneumonitis in patients with locally advanced
lung tumors: a comparison of volumetric modulated arc non-small-cell lung cancer treated with concurrent chemo-
therapy with three other delivery techniques. Radiother therapy and intensity-modulated radiotherapy. Radiat Oncol
Oncol 97(3):437–442. doi:10.1016/j.radonc.2010.09.027 12:35. doi:10.1186/1748-717X-5-35
Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K, Sura S, Gupta V, Yorke E, Jackson A, Amols H, Rosenzweig KE
Karasawa K, Hayakawa K, Niibe Y, Takai Y, Kimura T, (2008) Intensity modulated radiation therapy (IMRT) for
Takeda A, Ouchi A, Hareyama M, Kokubo M, Kozuka T, inoperable non-small-cell lung cancer: the Memorial Sloan-
Arimoto T, Hara R, Itami J, Araki T (2010) Stereotactic Kettering Cancer Center (MSKCC) experience. Radiother
body radiotherapy (SBRT) for operable stage i non-small- Oncol 87:17–23. doi:10.1016/j.radonc.2008.02.005
cell lung cancer: Can SBRT Be Comparable to Surgery? Int Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,
J Radiat Oncol Biol Phys (ePub 2010 Jul 15). doi: Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom
10.1016/j.ijrobp.2009.07.1751 AT, Christian MC, Gwyther SG (2000) New guidelines to
Otto K (2008) Volumetric modulated arc therapy: IMRT in a evaluate the response to treatment in solid tumor. European
single gantry arc. Med Phys 35:310–317. doi:10.1118/1. Organization for Research and Treatment of Cancer,
2818738 National Cancer Institute of the United States, National
Pisters KM (2000) The role of chemotherapy in early-stage Cancer Institute of Canada. J Natl Cancer Inst 92(3):
(stage I and II) resectable non-small-cell lung cancer. Semin 205–216. doi:10.1093/jnci/92.3.205
Radiat Oncol 10:274–279 Timmerman R, Paulus R, Galvin J, Michalski J, Straube W,
Ramsey C, Spencer K, Alhakeem R, Olive AL (2001) Leaf Bradley J, Fakiris A, Bezjak A, Videtic G, Johnstone D,
position error during conformal dynamic arc and intensity Fowler J, Gore E, Choy H (2010) Stereotactic body
modulated arc treatments. Med Phys 28:67–72 radiation therapy for inoperable early stage lung cancer.
Rebueno NC, Welsh J (2009) Exploring the feasibility of dose- JAMA 303:1070–1076. doi:10.1001/jama.2010.261
escalation with IMRT and the effects on normal tissue Ung YC et al (2000) Fusing 18-fluorodeoxyglucose (FDG)-
structures for thoracic malignancies [Abstract 2616]. Int J hybrid PET to CT images significantly alters treatment
Radiat Oncol Biol Phys 75:S458–S459. doi:10.1016/j.ijrobp. planning in the radical treatment of non-small-cell carci-
2009.07.1048 noma of the lung. Int J Radiat Oncol Biol Phys 52:339–350
Rengan R, Rosenzweig KE, Venkatraman E, Koutcher LA, Fox Wang S, Liao Z, Wei X, Liu HH, Tucker SL, Hu CS, Mohan R,
JL, Nayak R, Amols H, Yorke E, Jackson A, Ling CC, Cox JD, Komaki R (2006) Analysis of clinical and
Leibel SA (2004) Improved local control with higher doses dosimetric factors associated with treatment-related pneu-
of radiation in large-volume stage III non-small-cell lung monitis (TRP) in patients with non-small-cell lung cancer
cancer. Int J Radiat Oncol Biol Phys 60:741–747. doi: (NSCLC) treated with concurrent chemotherapy and three-
10.1016/j.ijrobp.2004.04.013 dimensional conformal radiotherapy (3D-CRT). Int J Radiat
Intensity-Modulated Radiation Therapy and Volumetric-Modulated Arc Therapy for Lung Cancer 713

Oncol Biol Phys 66:1399–1407. doi:10.1016/j.ijrobp.2006. primary lung cancers to radiotherapy by combined F-18
07.1337 FDG PET-CT imaging. Radiat Oncol 23:18. doi:10.1186/
Wang C, Luan S, Tang G, Chen DZ, Earl MA, Yu CX (2008) 1748-717X-2-18
Arc-modulated radiation therapy (AMRT): a single-arc Wulf J, Baier K, Mueller G, Flentje MP (2005) Dose-response
form of intensity-modulated arc therapy. Phys Med Biol in stereotactic irradiation of lung tumors. Radiother Oncol
53:6291–6303. doi:10.1088/0031-9155/53/22/002 77:83–87
Welsh R, Grills IS, Deraniyagala R, Kestin L, Baschnagel A, Yom SS, Liao Z, Liu HH, Tucker SL, Hu CS, Wei X, Wang X,
Mangona V, Ye H, Chmielewski G (2010) Lobectomy, Wang S, Mohan R, Cox JD, Komaki R (2007) Initial
wedge resection, or stereotactic radiotherapy (SBRT) for evaluation of treatment-related pneumonitis in advanced-
stage I non-small-cell lung cancer: Which treatment yields stage non-small-cell lung cancer patients treated with
the best outcome? [Abstract 1088]. Int J Radiat Oncol Biol concurrent chemotherapy and intensity-modulated radio-
Phys 78:S180. doi:10.1016/j.ijrobp.2010.07.438 therapy. Int J Radiat Oncol Biol Phys 68:94–102
Werner-Wasik M, Scott C, Graham ML, Smith C, Byhardt RW, Yorke ED, Jackson A, Rosenzweig KE, Braban L, Leibel SA,
Roach M III, Andras EJ (1999) Interfraction interval does Ling CC (2005) Correlation of dosimetric factors and
not affect survival of patients with non-small-cell lung radiation pneumonitis incidence for non-small-cell lung
cancer treated with chemotherapy and/or hyperfractionated cancer (NSCLC) patients in a recently completed dose-
radiotherapy: a multivariate analysis of 1076 RTOG escalation study. Int J Radiat Oncol Biol Phys 63:672–682.
patients. Int J Radiat Oncol Biol Phys 44:327–331. doi: doi:10.1016/j.ijrobp.2005.03.026
10.1016/S0360-3016(99)00031-0 Yu CX (1995) Intensity-modulated arc therapy with dynamic
Willner J, Jost A, Baier K, Flentje M (2003) A little to a lot or a multileaf collimation: an alternative to tomotherapy. Phys
lot to a little? an analysis of pneumonitis risk from dose– Med Biol 40:1435–1449. doi:10.1088/0031-9155/40/9/004
volume histogram parameters of the lung in patients with Yu CX, Tang G (2011) Intensity-modulated arc therapy:
lung cancer treated with 3D-conformal radiotherapy. Strah- principles, technologies and clinical implementation. Phys
lenther Onkol 179:548–556 Med Biol 56:R31–R54. doi:10.1088/0031-9155/56/5/R01
Wolff D, Stieler F, Welzel G, Lorenz F, Abo-Madyan Y, Mai S, Yu CX, Symons MJ, Du MN, Martinez AA, Wong JW (1995) A
Herskind C, Polednik M, Steil V, Wenz F, Lohr F (2009) method for implementing dynamic photon beam intensity
Volumetric modulated arc therapy (VMAT) vs. serial tomo- modulation using independent jaws and multileaf collimator.
therapy, step-and-shoot IMRT and 3D-conformal RT for Phys Med Bio 40:769–787. doi:10.1088/0031-9155/40/5/005
treatment of prostate cancer. Radiother Oncol 93:226–233. Zhang P, Hugo GD, Yan D (2008) Planning study comparison of
doi:10.1016/j.radonc.2009.08.011 real-time target tracking and four-dimensional inverse plan-
Wong CY, Schmidt J, Bong JS, Chundru S, Kestin L, Yan D, ning for managing patient respiratory motion. Int J Radiat
Grills I, Gaskill M, Cheng V, Martinez AA, Fink-Bennett D Oncol Biol Phys 72:1221–1227. doi:10.1016/j.ijrobp.2008.
(2007) Correlating metabolic and anatomic responses of 07.025
Image-Guided Robotic Stereotactic
Ablative Radiotherapy for Lung Tumors:
The CyberKnife
Billy W. Loo Jr. and Iris C. Gibbs

Contents Abstract
Stereotactic ablative radiotherapy (SABR) is a new
1 Introduction.............................................................. 715 paradigm in radiation therapy, achieving high rates
2 The CyberKnife Image-Guided Robotic SABR of local tumor control with low toxicity through very
System ....................................................................... 716 high dose intensity and conformity, and requiring
3 Fiducial Marker Placement.................................... 716 highly precise and accurate delivery. The Cyber-
Knife robotic radiosurgery system, originally
4 Patient Positioning and Stabilization .................... 717
designed for frameless intracranial radiosurgery
5 Treatment Planning................................................. 718 using automated image-guidance, has unique char-
6 Treatment Delivery and Image Guidance ............ 719 acteristics suitable for lung tumor SABR, including
dynamic tumor tracking capabilities. We review here
7 Clinical Results of CyberKnife SABR for Lung
Cancer ....................................................................... 720 the technical characteristics and clinical outcomes of
lung tumor SABR using the CyberKnife system.
8 Conclusions and Future Directions ....................... 723
References.......................................................................... 723

1 Introduction

Stereotactic ablative radiotherapy (SABR), also


called stereotactic body radiation therapy (SBRT), has
emerged as a promising treatment for early stage
non-small cell lung cancer, particularly for patients
unable to tolerate surgical resection, and possibly as an
alternative to surgery for some appropriately selected
patients. It has also proven useful for the treatment of
pulmonary oligometastases when an alternative to sur-
gical metastasectomy is desirable. High rates of local
tumor control, in some cases rivaling the historical
results of surgery, have been demonstrated with
acceptable toxicity and the practical advantage of a short
B. W. Loo Jr. (&)  I. C. Gibbs course of treatment. The CyberKnife image-guided
Department of Radiation Oncology, robotic radiosurgery system has unique technical char-
Stanford University and Cancer Institute,
acteristics that make it well suited for SABR of tumors
875 Blake Wilbur Drive: MC 5847,
Stanford, CA 94305-5847, USA that move with breathing, including lung tumors.
e-mail: BWLoo@stanford.edu This chapter reviews the qualities of the CyberKnife

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 715
DOI: 10.1007/174_2011_276, Ó Springer-Verlag Berlin Heidelberg 2011
716 B. W. Loo Jr. and I. C. Gibbs

is the software that integrates the functionality of the


hardware, including an inverse treatment planning
system for generating conformal targeting and normal
tissue avoidance plans, and the treatment delivery soft-
ware that analyzes the imaging data acquired during
treatment to determine the appropriate corrections to
apply to the couch and linac positions to maintain ste-
reotactic accuracy. A number of optional components
are available, including upgrades to the linac (with a
higher dose rate up to 1,000 MU/min) and the couch (full
six degree of freedom robotic positioning), an automatic
collimator changer, and a computer-controlled variable
aperture collimator. Of note, Monte Carlo dose calcu-
lation is available optionally, but as discussed below
Fig. 1 The CyberKnife image-guided robotic stereotactic should really be considered a requirement when treating
ablative radiotherapy system at Stanford University, with major
hardware components labeled with curative intent in regions of heterogeneous tissue
density such as in or around the lungs, or adjacent to air
cavities as in the paranasal sinuses.
platform for lung tumor SABR, and provides a The key elements of producing ideal treatment
summary of clinical data using this system specifically. plans and safe treatment delivery include: (1) proper
placement of fiducial markers; (2) proper patient
positioning and stabilization; (3) appropriate target
2 The CyberKnife Image-Guided delineation and margin design; (4) producing an
Robotic SABR System optimal dose distribution during treatment planning;
and (5) vigilant monitoring during treatment delivery.
The key distinguishing feature of the CyberKnife ste-
reotactic radiotherapy system is the seamless integration
of fully image-guided stereotactic localization, elimi- 3 Fiducial Marker Placement
nating the need for rigid anatomic fixation to achieve
precise and accurate radiation delivery. The CyberKnife Dynamic target tracking is an integral part of treating
was the first commercial platform to deliver modern tumors that move with breathing using the Cyber-
image-guided radiosurgery (IGRS), and remains the Knife. This process requires the implantation of
system with the greatest degree of automated image metallic fiducial markers in or around the tumor for
guidance. This capability has evolved from its original image-guided tracking. Markers may be delivered by
application to intracranial stereotactic radiosurgery a number of routes, including CT-guided percutane-
(SRS), to SABR of tumors in the body. Its unique ability ous needle placement, a bronchoscopic procedure, or
to dynamically track targets that move with breathing, even endovascular delivery, prior to the initial simu-
such as in the thorax and upper abdomen, differentiates lation (Kothary et al. 2009; Hong et al. 2010; Prévost
the CyberKnife from other commercially available et al. 2008; Anantham et al. 2007). The fiducial
image-guided platforms, which typically use either markers are ideally placed such that their projections
respiratory manipulation or respiratory gating in order to from the perspective of both of the in-room X-ray
manage breathing-induced motion. imagers are distinct, i.e., non-overlapping and well
The major hardware components of the system are: a separated (by about 1 cm) while still being in close
compact 6 MV linear accelerator mounted on a com- proximity to the lesion to be treated. Three markers
puter-controlled robotic manipulator, a pair of orthog- are sufficient for unique spatial localization, but in
onal X-ray sources and imaging panels, infrared practice 4–5 are often placed in case of loss or sub-
light-emitting diodes as external surface markers and an optimal placement of markers. Implantation and
optical camera that monitors their position, and a com- tracking of a single marker in the center of the tumor
puter-controlled patient couch (Fig. 1). Equally critical has also been described (Brown et al. 2009).
Image-Guided Robotic Stereotactic Ablative Radiotherapy 717

The main challenge in the use of fiducial markers is treatment system. Because the CyberKnife relies
the invasiveness of the implantation approaches. In entirely on image guidance for its accuracy, and
fact, the main acute toxicity encountered in CyberKnife because it adapts dynamically to intrafractional chan-
SABR of lung tumors is implantation-related pneu- ges in the target position, requirements for immobili-
mothorax. The CT-guided percutaneous approach in zation are far less stringent. In fact, given the relatively
particular carries the highest risk of pneumothorax, long treatment times with SABR using the CyberKnife,
although this is still a risk albeit small using broncho- less rigid immobilization is preferred: a higher priority
scopic approaches. Clinical studies of CyberKnife should be placed on patient comfort, which reduces the
SABR using percutaneous marker placement have likelihood of sudden large motions for which the sys-
reported 19–45% rates of any pneumothorax, and tem cannot compensate other than by interrupting the
3–26% rates of pneumothorax requiring temporary treatment. It makes more sense to think in terms of
chest tube placement (Kothary et al. 2009; Hong et al. patient position stabilization rather than immobiliza-
2010; Le et al. 2006; Collins et al. 2009; Pennathur et al. tion, as this is the more realistic and physiologic goal.
2009). These rates are similar to those of CT-guided A commonly used device for position stabilization is
percutaneous needle biopsies, and are likely related to a custom molded cushion consisting of a bag filled with
the general frailty of this patient population with severe foam beads, the shape of which is fixed upon evacua-
comorbidities such as chronic obstructive pulmonary tion of air from the bag after forming it around the
disease making them medically inoperable. patient (vacuum bag). The highest priority should be to
By contrast, Erasmus Medical Center investigators provide adequate support of the patient throughout the
found no pneumothorax associated with endovascular duration of treatment, and a full body-length cushion is
(pulmonary arterial) delivery of 87 vascular emboli- desirable. Maximum rigidity of the mold is unneces-
zation coils in 23 patients (Prévost et al. 2008), but sary, and can in fact be detrimental from the standpoint
cardiac arrythmia requiring pacemaker placement was of patient comfort and thus stability. For targets in the
observed in one patient after intravascular coil body, it is generally dosimetrically advantageous to
implantation in a subsequent study by the same group place the upper extremities in the overhead position.
(van der Voort van Zyp et al. 2009). Similarly, low It is important to provide adequate support to the upper
rates of 0–5.8% pneumothorax were found in patients extremities by building up an adequate mass of the
who underwent electromagnetic navigation broncho- cushion material such that the patient expends no
scopy or endobronchial ultrasound for endobronchial further effort to maintain that position. Fatigue and pain
implantation of various types of fiducial markers will impair positioning stability and may lead to
(Anantham et al. 2007; Harley et al. 2010; Schroeder undesirable treatment interruptions.
et al. 2010). Also, retention of implanted fiducial The CyberKnife is designed to use many non-
markers in lung tissue varies by marker type. For coplanar beams, which contributes to the highly
example, the Stanford group found that percutane- conformal plans achievable with this system. However,
ously implanted platinum endovascular embolization there are limits to the available beam directions based on
coils were much better retained than standard smooth geometrical constraints. When the patient is supine,
gold markers, and could successfully be tracked by beams are restricted to entering from approximately the
the CyberKnife after appropriate acceptance testing anterior hemisphere, because there is insufficient clear-
(Hong et al. 2010). Likewise, similar coils were much ance for the linac to be positioned under the patient to
better retained than linear fiducial markers implanted deliver beams from those directions. Similarly, there is a
bronchoscopically (Schroeder et al. 2010). more subtle asymmetry in beam access when treating
lateralized targets because the base of the robotic
manipulator is positioned on one side or the other of the
4 Patient Positioning couch, depending on the room configuration.
and Stabilization Therefore, while most patients are positioned supine,
for certain target locations, particularly very posterior or
Classically, SRS for brain tumors has required invasive lateral tumors, prone positioning can confer significant
fixation of the skull to a frame with a precisely known advantages in dose distribution given the available beam
spatial relationship relative to the isocenter of the access on the CyberKnife. Prone positioning permits
718 B. W. Loo Jr. and I. C. Gibbs

treatment of posterior thoracic lesions while avoiding a scan. During treatment delivery, the robotic manipu-
long path of entrance dose through the lungs that would lator follows the trajectory of the implanted fiducial
be required in the supine position (Ding et al. 2010). In markers, ensuring that there is no geometric miss of
addition, for very lateral tumors that would be on the the target. However, the dose to the surrounding
opposite side of the couch relative to the robotic normal structures can in principle be different from
manipulator when in the supine position, prone posi- that indicated by the static plan. If 4D CT simulation
tioning can increase the beam access and improve the is available, the treatment planning system can cal-
dose distribution by moving the target to the side of the culate the dose on all the phases of the 4D CT data set
robot. The use of a U-shaped cushion to support the face based on dynamic fiducial tracking, and deformably
without obstructing breathing may be used to facilitate propagate the doses to the reference phase, providing
comfort in that position. an estimate of the dynamic dose distribution. This
feature is an optional addition to the treatment plan-
ning system. It is also possible to optimize the beam
5 Treatment Planning weights based on organ motion over the phases of the
4D CT. However, this approach assumes that the
Treatment planning begins at simulation, where motion captured in the 4D CT scan (which effectively
patient positioning and image acquisition parameters represents a sampling of a single respiratory cycle in
can have a substantial impact on the quality of the time) is reproduced consistently during treatment,
plan and accuracy of treatment delivery subsequently. generally an overly optimistic assumption.
X-ray computed tomography (CT) is the fundamental A shortcoming of conventional dose calculation
imaging modality used for treatment planning. algorithms is their poor modeling of dose build-up and
Because of the non-coplanar beam arrangement, the penumbra from lateral electron scatter when radiation
total axial field of view of the scan (in the superior– beams traverse interfaces between materials of sub-
inferior direction) should be sufficiently long to stantially different density. The standard dose calcu-
include adequate tissue above and below the target lation algorithm in the CyberKnife treatment planning
volume for accurate calculation of attenuation of system uses a pencil beam (electronic path length,
oblique beams. In addition, because the image guid- EPL) model that produces accurate dose distributions
ance is based on comparison of the images acquired for targets in regions of homogeneous density such as
by the orthogonal in-room imagers with digitally the brain. However, it has significant inaccuracies
reconstructed radiographs (DRRs) generated from the when used in regions of sharp density gradients such as
corresponding projections through the CT data, in or around the lungs or air sinuses in the head. Monte
acquisition of the CT with thin cuts (1.25 mm or finer Carlo dose calculation, which models the interactions
slice spacing) is needed to produce high-resolution generated by individual photons to produce accurate
DRRs for optimal position and motion compensation. dose distributions when simulating many events, is
One distinguishing feature of the CyberKnife available as an optional addition to the treatment
treatment planning system is that it uses inverse planning system. When treating tumors in the lung, the
optimization. This allows simultaneous conformal discrepancy in calculated dose (covering specified
targeting and normal tissue avoidance, and also per- volume of the target) between the EPL and Monte
mits ‘‘dose painting’’, or simultaneously prescribing Carlo algorithms is typically 10–20% but can exceed
different doses to high- or low-risk regions, for 80% depending on the size and location of the specific
example corresponding to gross and microscopic target, with the largest discrepancies for small tumors
tumor extension. User selected parameters include the surrounded by air-filled lung parenchyma (Sharma
number and size of collimators, and exclusion of et al. 2010; Wilcox et al. 2010; van der Voort van Zyp
beams through specified structures. A typical SABR et al. 2010). The EPL calculation consistently over-
plan uses approximately 100–200 beams (Fig. 2). estimates the tumor coverage, but the degree of error is
The CyberKnife is designed to compensate for not generalizable or predictable without doing an
breathing-induced target motion by dynamic tracking, actual comparative calculation. Thus the Monte Carlo
as discussed in detail below. Of note, a tumor tracking module should be considered a requirement for
plan can be created from a static (breath-hold) CT definitive treatment of thoracic tumors.
Image-Guided Robotic Stereotactic Ablative Radiotherapy 719

Fig. 2 A typical lung tumor SABR treatment plan on the The prescription isodose line (green) conforms in all planes to
CyberKnife. The upper left panel shows the available beams the planning target volume (red), while lower isodose lines
(dark blue) as well as the beams used in the plan (light blue). avoid normal structures such as the esophagus and spinal canal

the original simulation. Subsequent changes in the


6 Treatment Delivery and Image patient’s position or breathing-induced target motion
Guidance are compensated for by adjustments to the linac
position using the robotic manipulator. The dynamic
Image guidance is in fact the key distinguishing fea- marker-based tumor tracking is then initiated (using the
ture of the CyberKnife treatment system. The degree Synchrony Respiratory Tracking System).
to which it is integrated into the delivery control Two separate imaging systems are employed in the
software and automated is what makes possible the tumor tracking schema (Schweikard et al. 2000,
dynamic tumor tracking feature. The CyberKnife is 2004). The first is the pair of X-ray imagers that
currently the only commercial platform that performs acquires orthogonal images of the fiducial markers,
dynamic tumor tracking. which are compared to the DRRs to obtain three-
At the beginning of treatment, in-room X-ray ima- dimensional localization of each of the markers as
ges are acquired and compared to the DRRs to calculate well as their center of mass. The image analysis
any required adjustment of the patient’s position. The software automatically extracts the projections of
initial corrections are applied by moving the couch to the markers from the images. Of note, the in-room
place the patient as closely as possible to the position of imagers produce intermittent static images rather than
720 B. W. Loo Jr. and I. C. Gibbs

continuous images. The second is an optical camera imagers, i.e., those located in the lung periphery at
that continuously monitors the position of three least 1.5 cm in size, and that do not overlap other
infrared light emitting diodes that are placed on the dense anatomical structures, such as the spine, dia-
external surface of the patient’s chest at the beginning phragm, and heart in the projection views. As such,
of the treatment session, and move as the patient marker-less tracking may be applicable to a minority
breathes. The trajectories of the external markers of lung tumors currently. Furthermore, its accuracy
follow the breathing cycle, including variations in the remains to be rigorously validated clinically, and
breathing pattern. The key to dynamic tracking with presently all of the publications on this subject pertain
this arrangement is the correlation of the intermittent to measurement of its performance in artificial set-
X-ray localization of the internal markers with the tings, although early clinical reports of favorable
continuous signal of the external markers. tumor control when using this method provide indi-
Prior to the initiation of treatment, a series of rect evidence of its accuracy (Brown et al. 2009).
10–15 X-ray images is acquired, timed such that the A typical lung tumor SABR plan is delivered in
internal marker coordinates from the various portions 60–90 min owing to the sequential delivery of
of the breathing cycle are all sampled, as determined approximately 100–200 non-coplanar beams. Options
by the signal from the optical camera. A correlation are available for a higher output linac with a dose rate
model is then built, which generates a continuously up to 1,000 MU/min, a treatment couch capable of
calculated position of the center of mass of the full 6 degree of freedom robotic positioning, and a
internal fiducials from the external marker coordi- variable aperture collimator, which together can
nates. It is this calculated/predicted position that the reduce treatment times by as much as 20–30%,
beam follows dynamically. making the treatment times comparable to those of
Intermittent X-ray images continue to be acquired other linac-based SABR treatment systems.
throughout the delivery, as often as at the beginning While the most unique strength of the CyberKnife
of every beam. Each new measurement of internal system is its automated image-guided motion com-
marker position is compared to the corresponding pensation, this in no way diminishes the importance
predicted position, and the correlation error is calcu- a highly skilled and attentive treatment team. A
lated. Treatment continues as long as the correlation potential pitfall is the temptation to allow the entire
error is below a user-defined threshold, generally treatment session to proceed automatically with
3 mm. This threshold is thus an approximate upper almost no user interaction. Because errors in auto-
bound to the tracking error of the system. If it is found mated image analysis, such as fiducial marker
to be exceeded, treatment stops, additional images are extraction, can and do occur, manual inspection of the
acquired, and if necessary, the correlation model is images displayed during the treatment and interrup-
rebuilt from the new images. Otherwise, the correla- tion for reimaging when necessary are crucial to
tion model is simply updated with the newest data ensure that treatment is delivered appropriately.
point. Gradual changes to the breathing pattern Ultimately, the treating physicians and therapy staff,
throughout treatment are accommodated in this way. rather than the technology itself, are the most
The clinical accuracy of the Synchrony system was important determinants of quality treatment and
assessed retrospectively in 44 patients, finding a technical accuracy.
maximum correlation model error of 2.5 mm
(standard deviation) (Hoogeman et al. 2009).
An optional marker-less tracking capability is also 7 Clinical Results of CyberKnife SABR
available for treatment of selected lung tumors for Lung Cancer
(the XSight Lung Respiratory Tracking System).
Tumor localization is accomplished using automated A large, rapidly growing literature has emerged in
real-time image segmentation of the in-room X-ray recent years documenting the high promise of SABR
images based on the contrast of the tumor itself. Thus for early lung cancer using a wide range of treatment
the system is best used for lesions with sufficient platforms. Notable for being a prospective coopera-
contrast in density from the surrounding anatomy to tive group trial in a uniform population of medically
be clearly visualized on both the in-room X-ray inoperable patients with peripherally located early
Image-Guided Robotic Stereotactic Ablative Radiotherapy 721

lung cancer, the Radiation Therapy Oncology Group possible treatment-related deaths, all in patients who
(RTOG) 0236 study demonstrated 98% local control had received prior or subsequent chemotherapy and
(within the primary tumor) and 87% local–regional two of three in patients who had received prior tho-
control (within the ipsilateral lobe, hilum, and medi- racic radiotherapy. Pulmonary toxicity was encoun-
astinum, consistent with the surgical definition of tered at doses 25 Gy or higher, mainly in patients
local control) at three years with an intensive regimen with central tumors or PTV greater than 50 mL (cm3),
of 60 Gy in three fractions (or approximately 54 Gy leading to the conclusion that 25 Gy single fraction
with proper heterogeneity corrections) (Timmerman SABR for small lung tumors is safe in properly
et al. 2010). Similarly, a prospective multi-institu- selected patients. No significant changes in pulmon-
tional study by the Nordic Study Group of SBRT ary function testing (PFT) were found in 17 patients
demonstrated 92% primary tumor control at three who had pre- and post-treatment PFT. With respect to
years (Baumann et al. 2009). tumor control, local control appeared to be better with
With respect to SABR using the CyberKnife spe- higher doses (91% at 1 year for doses C25 Gy) and
cifically, to date there have been 22 peer-reviewed primary lung cancer histology (as opposed to metas-
English language publications reporting the clinical tases). However, subsequent analysis of outcomes in
outcomes of lung tumor SABR with this platform, this series found the main predictor of local control
including updates of previously reported series with single fraction SABR to be tumor volume, with
(Brown et al. 2009; Le et al. 2006; Collins et al. 2009; excellent local control of tumors smaller than 12 mL
Pennathur et al. 2009; van der Voort van Zyp et al. but inadequate control of larger tumors in the dose
2009; Whyte et al. 2003; Nuyttens et al. 2006; Brown range tested: the Kaplan–Meier estimate of local
et al. 2007; Brown et al. 2007; Brown et al. 2007; control at 11 months was 100% for tumors \6 mL,
Collins et al. 2007; Muacevic et al. 2007; Pennathur 93% for tumors 6–12 mL, and 47% for tumors
et al. 2007; Brown et al. 2008; Coon et al. 2008; Ahn [12 mL (Brown et al. 2010). More recently, a pre-
et al. 2009; Pennathur et al. 2009; Brown et al. 2010; liminary analysis of a tumor volume-adapted dosing
Vahdat et al. 2010; van der Voort van Zyp et al. 2010; strategy in which small tumors (\12 mL) were trea-
van der Voort van Zyp et al. 2010; Unger et al. 2010). ted with single fractions of biologically effective dose
The majority of these comprise the experiences of five (BED) \100 Gy and larger tumors (C12 mL) were
major centers: Stanford University, Erasmus Medical treated with more dose intensive multi-fraction regi-
Center in Rotterdam, University of Pittsburgh, mens of BED C100 Gy found equally high 1-year
Georgetown University, and the CyberKnife Center local control rates of 91.4 and 92.5%, respectively in
of Miami. 83 patients with 97 tumors at a median follow-up of
In 2003, the first published report of lung tumor 13.5 months (Chang et al. 2009; Trakul et al. 2010).
SABR using the CyberKnife was of the preliminary Figure 3 shows an example of the treatment response
results and feasibility analysis of a prospective phase I following CyberKnife SABR of a peripheral lung
dose escalation trial conducted by Stanford University tumor.
and Cleveland Clinic that was in fact the first pub- Investigators from Erasmus Medical Center
lished North American clinical trial of SABR for lung reported a retrospective series of 70 patients with
cancer (Whyte et al. 2003). Twenty-three patients inoperable early stage peripheral NSCLC (39 T1
with primary NSCLC (15 patients) or single lung tumors; 31 T2 tumors) treated with either 45 or 60 Gy
metastases (8 patients) were treated in a single frac- in three fractions (1 treated to 36 Gy) (van der Voort
tion of 15 Gy (the starting dose level of the Phase I van Zyp et al. 2009). The median follow-up was 15
trial). In the early period of this study, motion man- months. There was a trend toward improved local
agement was achieved by respiratory breath-hold control with higher radiation dose, with 2 yr actuarial
technique or an early version of respiratory-tracking local control of 96% in those treated with 60 Gy
techniques that required very long treatment times. versus 78% in those treated with 45 Gy (P = 0.197).
The updated results of the completed phase I study at The four local recurrences were in patients with T2
Stanford University included 32 patients (20 NSCLC, tumors. Actuarial overall survival was 83 and 62% at
12 metastases) treated with 15, 20, 25, or 30 Gy in a first and second years, respectively, with 19 deaths
single fraction (Le et al. 2006). There were three during follow-up (6 from lung cancer, 13 from
722 B. W. Loo Jr. and I. C. Gibbs

Fig. 3 Peripherally located


stage I NSCLC in a medically
inoperable patient, treated
with CyberKnife SABR to
a dose of 50 Gy in four
fractions. There was a
complete response with minor
tissue retraction surrounding
the implanted markers
(arrow), and no evidence
of disease 26 months after
treatment

intercurrent illness), yielding cause-specific survival was 24 months. There was a statistically significantly
rates of 94 and 86% at 1 and 2 years, respectively. longer time to local progression with the higher dose.
Late toxicities included grade 3 pneumonitis in three University of Pittsburgh investigators are also coor-
patients, and grade 3 thoracic pain in four patients dinating a multi-institutional phase II clinical trial of
with lesions near the chest wall. An analysis by the CyberKnife SABR for medically inoperable early
same institution of a subgroup of 38 octagenarians lung cancer, which continues to accrue patients
with stage I NSCLC found 100% 2 year local control (ClinicalTrials.gov 2008).
and 1- and 2-year overall survival of 65 and 44%, Georgetown University investigators reported a
respectively, which are favorable results in this series of 20 medically inoperable patients with small
especially frail patient population (van der Voort van peripheral stage I NSCLC treated with CyberKnife
Zyp et al. 2010). In addition, a quality of life analysis SABR to an average dose of 53 Gy (range 42–60 Gy)
of 39 patients on a prospective phase II trial of in 3 fractions over a 3–11 day period (Vahdat et al.
4860 Gy in 36 fractions found maintenance of quality 2010). The mean tumor volume in this series was
of life and significant improvement in emotional 10 mL (range 4–24 mL). With a median follow-up of
functioning after SABR along with 97% local control 43 months, the 2 year actuarial survival was 90% and
and 62% overall survival at 2 years and no severe local control was 95%. No regional and three distant
([grade 3) treatment-related toxicity (van der Voort recurrences were observed. Chest wall discomfort
van Zyp et al. 2010). occurred in 8 of 12 patients with tumors near the
The University of Pittsburgh group reported their pleura and 1 case of subacute grade 3 pneumonitis
retrospective experience treating 100 patients, 46 with was encountered in a patient who had received radi-
primary NSCLC, 35 with locally recurrent tumors ation concurrently with gefitinib (Collins et al. 2009).
after prior therapy, and 19 with pulmonary metastasis They also reported the outcomes of 20 patients with
(Pennathur et al. 2009). The majority (72 patients) large hilar tumors (median gross tumor volume
were treated with 20 Gy in a single fraction, while 28 73 mL) treated more modest doses of 30–40 Gy in
received 60 Gy in three fractions. With a median five fractions, finding an estimated 1-year local
follow-up of 20 months, median time to local pro- control of 63%, suggesting a palliative rather than
gression was 22 months and median overall survival definitive benefit of such regimens in this setting
Image-Guided Robotic Stereotactic Ablative Radiotherapy 723

(Unger et al. 2010). One patient with gross endo- tracking of tumors to reduce the need for invasive
bronchial tumor invasion developed a fatal fistula marker placement. Rigorous clinical and technical
after treatment. evaluation will be important to validate the accuracy
The CyberKnife Center of Miami group reported a of these enhancements.
retrospective series of 31 patients with Stage IA or IB
NSCLC with tumors ranging in volume from 0.6 to
71 mL treated to doses of 60–67.5 Gy in 3–5 frac-
tions (Brown et al. 2009). After a median follow-up
References
time of 27.5 months, actuarial local control rates of
93.2 and 85.8% were observed at 1 and 4.5 years, Ahn SH, Han MS, Yoon JH et al (2009) Treatment of stage I
non-small cell lung cancer with CyberKnife, image-guided
respectively, and the overall survival was 93.6 and robotic stereotactic radiosurgery. Oncol Rep 21:693–696
83.5% at 1 and 4.5 years, respectively. There were no Anantham D, Feller-Kopman D, Shanmugham LN et al (2007)
observed grade 3 or higher toxicities. Electromagnetic navigation bronchoscopy-guided fiducial
In summary, the clinical outcomes of CyberKnife placement for robotic stereotactic radiosurgery of lung
tumors: a feasibility study. Chest 132:930–935
SABR of pulmonary tumors are very consistent with Baumann P, Nyman J, Hoyer M et al (2009) Outcome in a
the promising results reported in the broader literature prospective Phase II trial of medically inoperable Stage I
of SABR for lung tumors. non-small cell lung cancer patients treated with stereotactic
body radiotherapy. J Clin Oncol 27:3290–3296
Brown JM, Diehn M, Loo BW (2010) Stereotactic ablative
radiotherapy should be combined with a hypoxic cell
8 Conclusions and Future Directions radiosensitizer. Int J Radiat Oncol Biol Phys 78:323–327
Brown WT, Wu X, Amendola B et al (2007a) Treatment of early
SABR has been shown to be effective and feasible for non-small cell lung cancer, stage IA, by image-guided robotic
stereotactic radioablation–CyberKnife. Cancer J 13:87–94
the treatment of medically inoperable early stage Brown WT, Wu X, Fayad F et al (2007b) CyberKnife
NSCLC, and the CyberKnife system employs novel radiosurgery for stage I lung cancer: results at 36 months.
solutions to some of the technical challenges involved Clin Lung Cancer 8:488–492
in these complex treatments. Treatments of high Brown WT, Wu X, Wen BC et al (2007c) Early results of
CyberKnife image-guided robotic stereotactic radiosurgery
biologically effective doses (e.g., 60 Gy in three for treatment of lung tumors. Comput Aided Surg 12:253–261
fractions) have yielded excellent local control results, Brown WT, Wu X, Fayad F et al (2009) Application of robotic
particularly for smaller, peripheral tumors. Additional stereotactic radiotherapy to peripheral stage I non-small cell
studies are needed to optimize the dose for large and lung cancer with curative intent. Clin Oncol (R Coll Radiol)
21:623–631
centrally located tumors. Given the favorable results Brown WT, Wu X, Fowler JF et al (2008) Lung metastases
that are competitive with results historically observed treated by CyberKnife image-guided robotic stereotactic
after surgery for these tumors, ongoing trials aim to radiosurgery at 41 months. South Med J 101:376–382
study this treatment modality in operable patients, Chang CN, Zhou LY, MacFarlane G et al (2009) Excellent
early local control with tumor volume adapted dosing of
including multi-center cooperative group trials being stereotactic body radiation therapy for pulmonary tumors.
conducted by the RTOG and the Japan Clinical J Thorac Oncol 4:S938–S939
Oncology Group (JCOG). In addition to these, the ClinicalTrials.gov (2008) CyberKnife radiosurgical treatment
international Lung Cancer STARS (Stereotactic of inoperable early stage non-small cell cancer. http://www.
clinicaltrials.gov/show/NCT00643318. Accessed 1 Mar 2011
Radiotherapy vs. Surgery) trial, coordinated by the ClinicalTrials.gov (2009) International randomized study to
M.D. Anderson Cancer Center and sponsored by compare CyberKnife stereotactic radiotherapy with surgical
Accuray, Inc., will study in a randomized fashion the resection in stage I non-small cell lung cancer (STARS).
comparative effectiveness of fractionated SABR http://www.clinicaltrials.gov/show/NCT00840749. Accessed
1 Mar 2011
(nominally 60 Gy in three fractions for peripheral Collins BT, Erickson K, Reichner CA et al (2007) Radical
lesions, and 60 Gy in four fractions for central stereotactic radiosurgery with real-time tumor motion
lesions) using the CyberKnife system specifically tracking in the treatment of small peripheral lung tumors.
versus surgical lobectomy for stage I NSCLC Radiat Oncol 2:39
Collins BT, Vahdat S, Erickson K et al (2009) Radical
(ClinicalTrials.gov 2009). In parallel, technological cyberknife radiosurgery with tumor tracking: an effective
improvements in hardware and software should lead treatment for inoperable small peripheral stage I non-small
to faster delivery times and improved marker-less cell lung cancer. J Hematol Oncol 2:1
724 B. W. Loo Jr. and I. C. Gibbs

Coon D, Gokhale AS, Burton SA, Heron DE, Ozhasoglu C, Prévost J-BG, Nuyttens JJ, Hoogeman MS, Pöll JJ, van Dijk
Christie N (2008) Fractionated stereotactic body radiation LC, Pattynama PMT (2008) Endovascular coils as lung
therapy in the treatment of primary, recurrent, and meta- tumour markers in real-time tumour tracking stereotactic
static lung tumors: the role of positron emission tomogra- radiotherapy: preliminary results. Eur Radiol 18:1569–1576
phy/computed tomography-based treatment planning. Clin Schroeder C, Hejal R, Linden PA (2010) Coil spring fiducial
Lung Cancer 9:217–221 markers placed safely using navigation bronchoscopy in
CyberKnife radiosurgical treatment of inoperable early stage inoperable patients allows accurate delivery of CyberKnife
non-small cell cancer. Accessed March 1, 2011. http://www. stereotactic radiosurgery. J Thorac Cardiovasc Surg 140:
clinicaltrials.gov/show/NCT00643318 1137–1142
Ding C, Chang C-H, Haslam J, Timmerman R, Solberg T Schweikard A, Glosser G, Bodduluri M, Murphy MJ, Adler JR
(2010) A dosimetric comparison of stereotactic body (2000) Robotic motion compensation for respiratory movement
radiation therapy techniques for lung cancer: robotic versus during radiosurgery. Comput Aided Surg 5:263–277
conventional linac-based systems. J Appl Clin Med Phys/ Schweikard A, Shiomi H, Adler J (2004) Respiration tracking
Am Coll Med Phys 11:3223 in radiosurgery. Med Phys 31:2738–2741
Harley DP, Krimsky WS, Sarkar S, Highfield D, Aygun C, Sharma SC, Ott JT, Williams JB, Dickow D (2010) Clinical
Gurses B (2010) Fiducial marker placement using endo- implications of adopting Monte Carlo treatment planning
bronchial ultrasound and navigational bronchoscopy for for CyberKnife. J Appl Clin Med Phys/Amer Coll Med Phys
stereotactic radiosurgery: an alternative strategy. Ann 11:3142
Thorac Surg 89:368–373 Discussion 73-4 Timmerman R, Paulus R, Galvin J et al (2010) Stereotactic
Hong JC, Yu Y, Rao AK et al (2010) High retention and safety body radiation therapy for inoperable early stage lung
of percutaneously implanted endovascular embolization cancer. JAMA 303:1070–1076
coils as fiducial markers for image-guided stereotactic Trakul N, Harris J, Dieterich S et al (2010) Volume adapted
ablative radiotherapy of pulmonary tumors. Int J Radiat dosing in stereotactic ablative radiotherapy of lung tumors.
Oncol Biol Phys, in press, published online ahead of print Int J Radiat Oncol Biol Phys 78(3 suppl 1):S179
August 2010 Unger K, Ju A, Oermann E, et al. (2010) CyberKnife for hilar
Hoogeman M, Prévost J-B, Nuyttens J, Pöll J, Levendag P, lung tumors: report of clinical response and toxicity.
Heijmen B (2009) Clinical accuracy of the respiratory J Hematol Oncol 3:39–45
tumor tracking system of the cyberknife: assessment by Vahdat S, Oermann EK, Collins SP et al (2010) CyberKnife
analysis of log files. Int J Radiat Oncol Biol Phys 74: radiosurgery for inoperable stage IA non-small cell lung
297–303 cancer: 18F-fluorodeoxyglucose positron emission tomog-
International randomized study to compare CyberKnife stereo- raphy/computed tomography serial tumor response assess-
tactic radiotherapy with surgical resection in Stage I ment. J Hematol Oncol 3:6
non-small cell lung cancer (STARS). (Accessed March 1, van der Voort van Zyp NC, Prévost J-B, Hoogeman MS et al
2011, at http://www.clinicaltrials.gov/show/NCT00840749.) (2009) Stereotactic radiotherapy with real-time tumor
Kothary N, Heit JJ, Louie JD et al (2009) Safety and efficacy of tracking for non-small cell lung cancer: clinical outcome.
percutaneous fiducial marker implantation for image-guided Radiother and Oncol J Eur Soc Ther Radiol and Oncol
radiation therapy. J Vasc Interv Radiol: JVIR 20:235–239 91:296–300
Le Q-T, Loo BW, Ho A et al (2006) Results of a phase I dose- van der Voort van Zyp NC, Hoogeman MS, van de Water S
escalation study using single-fraction stereotactic radiother- et al (2010a) Clinical introduction of Monte Carlo treatment
apy for lung tumors. J Thorac Oncol 1:802–809 planning: a different prescription dose for non-small cell
Muacevic A, Drexler C, Wowra B et al (2007) Technical lung cancer according to tumor location and size. Radiother
description, phantom accuracy, and clinical feasibility for and Oncol J Eur Soc Ther Radiol Oncol 96:55–60
single-session lung radiosurgery using robotic image-guided van der Voort van Zyp NC, Prévost J-B, van der Holt B et al
real-time respiratory tumor tracking. Technol Cancer Res (2010b) Quality of life after stereotactic radiotherapy for
Treat 6:321–328 stage I non-small cell lung cancer. Int J Radiat Oncol Biol
Nuyttens JJ, Prévost J-B, Praag J et al (2006) Lung tumor Phys 77:31–37
tracking during stereotactic radiotherapy treatment with the van der Voort van Zyp NC, van der Holt B, van Klaveren RJ,
CyberKnife: Marker placement and early results. Acta Pattynama P, Maat A, Nuyttens JJ (2010c) Stereotactic body
Oncol 45:961–965 radiotherapy using real-time tumor tracking in octogenar-
Pennathur A, Luketich JD, Burton S et al (2007) Stereotactic ians with non-small cell lung cancer. Lung Cancer 69:
radiosurgery for the treatment of lung neoplasm: initial 296–301
experience. Ann Thorac Surg 83:1820–1824 discussion 4-5 Whyte RI, Crownover R, Murphy MJ et al (2003) Stereotactic
Pennathur A, Luketich JD, Heron DE et al (2009a) Stereotactic radiosurgery for lung tumors: preliminary report of a phase I
radiosurgery for the treatment of lung neoplasm: experi- trial. Ann Thorac Surg 75:1097–1101
ence in 100 consecutive patients. Ann Thorac Surg 88: Wilcox EE, Daskalov GM, Lincoln H, Shumway RC, Kaplan
1594–1600 discussion 600 BM, Colasanto JM (2010) Comparison of planned dose
Pennathur A, Luketich JD, Heron DE et al (2009b) Stereotactic distributions calculated by Monte Carlo and Ray-Trace
radiosurgery for the treatment of stage I non-small cell lung algorithms for the treatment of lung tumors with cyberknife:
cancer in high-risk patients. J Thorac Cardiovasc Surg a preliminary study in 33 patients. Int J Radiat Oncol Biol
137:597–604 Phys 77:277–284
Advances in Radiation Oncology
of Lung Cancer
Deepak Khuntia and Minesh P. Mehta

Contents Abstract
A variety of advanced radiation technologies are
1 Basics of Tomotherapy............................................ 725 currently available that have changed the way we
1.1 Helical Delivery......................................................... 725 practice Radiation Oncology. In this chapter, the
1.2 Planning Parameters .................................................. 726
1.3 Image Guidance......................................................... 727
authors will review one such technology, Helical
tomotherapy. Basic operations and planning, appli-
2 Clinical Application................................................. 728
cations in stereotactic body radiotherapy, and
2.1 Stereotactic Body Radiotherapy for NSCLC ........... 728
2.2 Unresectable NSCLC ................................................ 728 locally advanced lung cancer, as well as devel-
2.3 Hippocampal Sparing Whole Brain Radiation ......... 730 oping and future applications in hippocampal
3 Future Directions..................................................... 730 sparing whole brain radiation, adaptive radiation,
3.1 Adaptive Radiotherapy and theragnostic radiation delivery will also be
and Real-Time Planning............................................ 730 discussed.
4 Conclusion ................................................................ 732
References.......................................................................... 732
1 Basics of Tomotherapy

1.1 Helical Delivery

Tomotherapy is the ‘‘slice-by-slice’’ delivery of inten-


sity modulated radiation therapy, originally pioneered
by Mark Carol as ‘‘serial tomotherapy’’, and com-
mercialized as the NOMOS system. Helical tomo-
therapy, which replaces the ‘‘serial slice-by-slice
Disclosures Dr. Khuntia serves on the advisory board of
Radion Global and has received the speakers honorarium delivery’’ (which requires multiple couch position
for Tomotherapy. Dr. Mehta serves as a consultant for adjustments), represents a substantial degree of
Tomotherapy, and holds stock options in Tomotherapy.
sophisticated refinement over this approach, by
D. Khuntia (&) utilizing helical delivery on a couch in continuous
Western Radiation Oncology, slow-motion, and is currently commercially available
125 South Drive, Mountain View, as Tomotherapy Hi-Art and Tomotherapy HD
CA 94040, USA (Tomotherapy, Inc., Madison, WI). This represents
e-mail: khuntia@wradonc.com
just one of several image guided radiotherapy (IGRT)
M. P. Mehta delivery devices available in the market place. To-
Northwestern University,
676 N St. Clair, Suite 1800, Chicago, motherapy, however, differentiates itself from the
IL 60611, USA other devices wherein it is a dedicated IGRT-IMRT

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 725
DOI: 10.1007/174_2011_277, Ó Springer-Verlag Berlin Heidelberg 2011
726 D. Khuntia and M. P. Mehta

Fig. 1 Primary components of a helical tomotherapy unit


(courtesy of Rockwell Mackie, PhD)

Fig. 2 Helical Tomotherapy Hi-Art


system incorporates megavoltage CT for IGRT, and
delivers radiation with multiple coplanar rotational
radiation beamlets, and the ability to collect exit dose
from the patient, mapped back to the daily megavolt-
age CT imaging allows precise documentation and
calculation of the daily delivered dose (Mackie et al.
1999, 2003). The technology of tomotherapy dates
back to the 1990s with the original Peacock System
(Nomos, Sewickley, PA) where rotational delivery is
accomplished using a fan beam generated from fast-
moving, actuator-driven multileaf collimators (MLC).
This technology has evolved into what is now known
as Tomotherapy Hi-Art and Tomotherapy HD (Helical Fig. 3 Multi-leaf collimator within gantry of a Tomotherapy
and Direct). Tomotherapy Direct refers to the ability to Hi-Art system. Each MLC has an incredibly fast transit time of
deliver radiation with limited or fixed gantry positions 20 ms. The rapid rotation and modulations allow for conformal
radiation delivery
emulating a 3D plan or a forward planned IMRT.
With the current Tomotherapy Hi-Art device, the
MLC modulates the beam in the range of 300 cm/s 1.2 Planning Parameters
which allows incredible modulation of the beam,
exceeding any currently available commercially Each Tomotherapy system includes its own proprie-
available radiation technology (Figs. 1, 2, 3). The tary treatment planning system. The system, as a
beam delivers radiation through a ring gantry system, whole, is one of the most forgiving planning systems
similar to a diagnostic helical spiral CT scanner. and also one of the easiest to learn. Treatment plan-
There are multiple advantages to a ring gantry system, ning is based primarily on three parameters: field
including increased stability, faster rotation, simple width, pitch, and modulation factor. Further, each
coplanar delivery, and no possibility of rotational col- rotation is divided into 51 separate beam projections
lisions. Further, because of the intense modulation with each projection having its own leaf opening and
capable with this system, a single low energy (6 MV) is closing pattern which transcends over seven degrees
sufficient. This allows for less neutron contamination of arc. The first planning parameter is field width.
and less shielding requirements. Field width can have a significant impact on both
Advances in Radiation Oncology of Lung Cancer 727

conformality and treatment time. Specifically, field


width refers to the width of each slice of delivery
along the Y dimension of the 85-cm field length. Field
sizes of 1, 2.5, and 5 cm can be used; 1-cm field sizes
will allow for significant conformality when high
precision is needed [such as in stereotactic body
radiotherapy (SBRT)], but comes at the expense of
treatment time. A very large treatment field (such as
total marrow irradiation) may be better treated with a
5-cm field width.
Pitch refers to the field width the couch moves per
rotation. For example, if the field width is 5 cm and the
pitch is 0.5, the table will move 2.5 cm per rotation. In
other words, pitch refers to how loose or tight the
spiral of radiation delivery is. As one decreases the
pitch, there could be an increase in the number of Fig. 4 Megavoltage (MVCT) taken prior to treatment delivery
rotations that could increase the treatment time. for a stereotactic lung radiotherapy delivery of 12 Gy in a
However a short pitch may allow for improved single fraction. An additional four fractions will be delivered.
The grey box represents the planning kilovoltage CT with the
resolution in the dose distribution, as the spiral is tight. purple boxes representing the daily MVCT image. Adjustments
Finally, modulation factor is the third treatment are made to endure that the patient is setup in a similar position
planning parameter. This refers to the maximum leaf as the planning scan
opening time divided by the average leaf opening
time of the MLC. A high-modulation factor allows
more flexibility in the leaf opening time, resulting in be incorporated which will yield less normal tissue
more conformal plans, but can come at the expense of dose. Further, if there is a dramatic change in a
longer treatment delivery times. patient’s anatomy, this will allow the physician to
identify the changes earlier and apply a new radiation
plan if warranted (Siker et al. 2006).
1.3 Image Guidance The changes seen on MVCT can vary based on
the disease site being treated (Schubert et al. 2009).
Image-guided radiotherapy (IGRT) uses online Head and neck and brain treatments have smaller
imaging just prior to treatment delivery to improve shifts than lung, for example, as there tends to be
the probability that the patient is positioned accurately little motion with brain/neck treatments as these are
and reproducibly for every treatment fraction fixed structures and have very good immobilization.
(Dawson and Jaffray 2007). Currently, different IGRT The lung, however, can have considerable motion
systems use portal films, electronic portal images, and the currently available immobilization devices
infrared cameras, conebeam CT, radiofrequency cannot restrict motion to the same degree as in the
beacons via implanted fiducials, ultrasound, or MVCT head and neck. Therefore, image guidance is critical
(as is the case with Tomotherapy), and MR imaging in the chest, and becomes even more critical when
on-board is being investigated. Typically, the stan- hypofractionated schedules are employed. Schubert
dard practice has involved checking portal films et al. (2009) found that variations in systemic errors
approximately once in every five fractions. This and the magnitude of random errors were higher for
makes it difficult to see variations compared to the lung cancer than any other disease site. As tomo-
planning session which may be a result of human therapy is a fully integrated IGRT system, there is
error in setup, patient anatomy changing due to considerable advantage in this technology in man-
weight loss or edema, or even changes in the tumor. aging thoracic tumors. Further, it causes very low
By using daily MVCT for image guidance (Fig. 4), radiation exposure to the patient (1.5 cGy) compared
there is less uncertainty in patient positioning, so to other 3D IGRT techniques such as conebeam CT
tighter planning target volume (PTV) expansions can (Shah et al. 2008).
728 D. Khuntia and M. P. Mehta

2 Clinical Application

Tomotherapy is uniquely qualified as a premier


radiation delivery device for patients with lung
cancer. As described below, there is a changing par-
adigm in lung cancer, focusing on dose-escalation,
including the approach of dose-per-fraction escala-
tion, based on biologic rationale, and made possible
by a changing technological landscape. In the 3D
conformal radiotherapy era, Radiation Oncologists
were limited to moderate doses of radiation due to the
lack of conformality with these techniques. Modern
delivery techniques, especially as available with
Tomotherapy, have allowed safe dose escalation that Fig. 5 The figure above represents the dose escalation model
has resulted in improvements in local control, survival described by Mehta and colleagues. On the Y-axis, the tumor
and reduced toxicity (Adkison et al. 2008). control probability is represented while the X-axis represents
time. As can be seen, maximum TCP is realized at 25 fractions
(5 weeks). (Mehta and Scrimger et al. 2001)

2.1 Stereotactic Body Radiotherapy


for NSCLC
mean tumor regression was 72% and all the patients in
Stereotactic Body Radiotherapy (SBRT) refers to the original series were locally controlled with no grade
precise 3D volume positioning of targets outside the two or higher pulmonary toxicity. Others have found
brain, taking motion into account, to deliver extre- similar local control rates in excess of 90% using SBRT
mely high doses of radiation conformally in five or techniques for unresectable lung cancer (Nagata et al.
fewer fractions. In order to successfully deliver SBRT 2005; Timmerman et al. 2006; Yoon et al. 2006).
to lung tumors, proper selection is mandatory.
Generally, these tumors must be peripheral in location
as clinical trials have shown a higher incidence of life 2.2 Unresectable NSCLC
threatening toxicity for central tumors (though other
fractionation regimens are being investigated for this It has been generally accepted that a dose-response
group) (Timmerman et al. 2006; Chi et al. 2011). relationship exists for non-small cell lung cancer
Because lung tumors can move up to 5 cm (especially based on several prospective clinical trials and data-
for tumors near the diaphragm), it is important that a base analyses (Perez et al. 1980; Kong et al. 2005;
method exists to limit this motion or track the tumor Belderbos et al. 2006; Yuan et al. 2007). However,
and gate treatment delivery. Tomotherapy accom- until recently, safe delivery of significantly escalated
plishes this by immobilizing the patient (typically in doses was difficult to achieve. There have been at
an abdominal compression device) and planning the least two helical tomotherapy-based series in unre-
patient using a 4D CT scan and targeting the motion sectable locally advanced NSCLC that have shown
envelope. MVCT is used to ensure that the tumor is initial promise for this difficult disease. Mehta and
adequately targeted during treatment. colleagues described a hypofractionated dose-per-
One of the earliest SBRT experiences using helical fraction escalation model keeping the number of
tomotherapy was reported by Hodge and colleagues at fractions constant (25) in order to combat accelerated
the University of Wisconsin (Hodge et al. 2006). In their repopulation which can retard the gains from dose
series, patients received a total of 60 Gy in five fractions escalation associated with treatment prolongation
over a ten day period. The PTV was defined as the (Mehta et al. 2001). Based on the biological param-
motion envelope plus a 6 mm expansion for microscopic eters utilized, and as can be seen from Fig. 5, the
extension and setup error with daily MVCT IGRT. The maximal tumor control probability is likely to be
Advances in Radiation Oncology of Lung Cancer 729

Fig. 6 Typical dose escalation plan in a patient treated on the low doses are realized to the esophagus and lung with this plan
UW phase I dose escalation trial. The dose volume histogram is (Adkison et al. 2008)
shown below. This patient received 3 Gy 9 25 fractions. Very

achieved when treatment is delivered over 25 frac- Song et al. (2010) retrospectively reviewed 37
tions, permitting one to utilize this schedule as a NSCLC patients treated with hypofractionated radiation
baseline and escalating beyond it by increasing frac- to 60-70.4 Gy with 2-2.4 Gy per fraction to the gross
tional dose, rather than the number of fractions. disease delivered with helical tomotherapy, in con-
The phase I study reported by Adkison et al. junction with concurrent chemotherapy. The authors
(2008) from the University of Wisconsin binned reported incredibly high 2-year local control and sur-
patients into separate ‘‘dose-volume-risk’’ bins, and vival rates of 63 and 56%, respectively. Acute esopha-
escalated fractional doses from 2.28 to 3.62 Gy per gitis grade 3 or higher was seen in five patients and grade
fraction 9 25 fractions. Patients were placed into a 3 or higher pneumonitis was seen in seven patients. Four
variety of ‘‘Bins’’ based on predicted mean dose to the treatment-related deaths were reported. A multivariate
lung and esophagus (Fig. 6). At the time of initial risk analysis demonstrated an association between the
reporting in 46 patients, no grade three or higher volume of lung receiving low doses of radiotherapy and
toxicities had occurred. In this study, the use of the development of pulmonary toxicity, and the authors
concurrent chemotherapy was prohibited though non- advocated that no more than 60% of the contralateral
gemcitabine containing regimens which were allowed lung volume should receive 5 Gy or higher dose. The
neoadjuvantly or adjuvantly. An impressive 47% patient cohort included in their report had very advanced
2-year OS was seen, despite having over 80% of disease, including supraclavicular bulky N3 adenopathy
patients as Stage III or higher disease. with poor performance status. The two studies also
730 D. Khuntia and M. P. Mehta

Fig. 7 Typical hippocampal whole brain radiation therapy plan. Here, the goal is to keep the mean dose to the hippocampus under
10 Gy and preferably under 7–8 Gy. This is best accomplished with helical tomotherapy

differed in their GTV to CTV expansion margins, with compartments, and possibly reduce the likelihood of
the Song study utilizing more generous expansion cognitive decline.
margins. Regardless, the control rates were impressive, Considerable work has been done over the last
and this is clearly a group of patients that likely could not several years attempting to delineate the feasibility
be treated safely with 3D techniques. of hippocampal sparing WBRT. Investigators have
shown that the likelihood of isolated recurrences
within the hippocampus is small (Gondi et al. 2010).
2.3 Hippocampal Sparing Whole Brain Further, atlases have been created to help standard-
Radiation ize the contouring of the hippocampus so that this
technique might be broadly, but reproducibly applied
Given the fact that a significant proportion of (Gondi et al. 2010). Patients requiring prophylactic
patients with lung cancer develop brain metastasis, cranial radiation or those with very good perfor-
whole brain radiation therapy (WBRT) is an mance status requiring WBRT may have the most to
important treatment for many of these patients gain from such an approach. Unfortunately, the
(Khuntia et al. 2006). WBRT has been shown to ability to deliver such exquisite plans where the
improve local control as well as delay decline in whole brain receives 30 Gy or more while the hip-
neurocognitive function (NCF) (Aoyama et al. pocampus receives mean doses of 10 Gy or less is
2007). However, there may be adverse effects on very difficult to accomplish. Helical tomotherapy has
NCF associated with WBRT and prospective efforts been shown to be able to accomplish some of the
are being made to minimize these (DeAngelis et al. most conformal radiation plans in this setting
1989; Chang et al. 2009) The RTOG recently com- (Fig. 7).
pleted a study (RTOG 0614) evaluating the drug
memantine to preserve cognitive function in patients
undergoing WBRT, and results are awaited. A cur- 3 Future Directions
rent RTOG trial, RTOG 0933, is evaluating the use
of IMRT to reduce the dose to the peri-hippocampal 3.1 Adaptive Radiotherapy
stem cell compartments involved in neurogenesis and Real-Time Planning
essential for memory functions (Monje et al. 2002;
Mizumatsu et al. 2003; Monje et al. 2003). By Adaptive radiotherapy refers to the ability to change
reducing the dose to the hippocampus, it may be the radiation delivery based on anatomic variation in
possible to preserve these radiosensitive stem cell a patient from one day to another. This has
Advances in Radiation Oncology of Lung Cancer 731

Fig. 8 The image on the left shows the treatment planning dramatic reduction in tumor (now outlined in blue with the
volume of a patient with small cell lung cancer outlined in red. original volume still in red.) Tomotherapy allows for
After just 1 week of radiation (15 Gy of radiation in ten re-planning to be accomplished from the daily MVCT so that
fractions, with twice daily treatments), there has been a a new plan can be delivered the following day

particular importance in patients where the external situations. Tomotherapy is therefore uniquely quali-
anatomy changes (such as a patient that has lost a lot fied to develop adaptive radiation therapy protocols
of weight during the course of treatment) or even in given its image guidance and treatment planning
patients that have rapidly responding tumors such as software capabilities.
small cell lung cancer (Fig. 8). Tomotherapy differs Further, this technology will allow for biologi-
from other delivery devices in that the image guided cally adapted radiotherapy, tailored to the response
MVCT images can be used for replanning. This biology of a patient’s tumor, a process referred to as
application, known as Planned Adaptive, is proprie- theragnostic radiotherapy. For example, it would be
tary to Tomotherapy. Further, investigations are quite possible to develop dose-prescription models
underway that will allow the use of advanced con- that track early alterations in resistance processes
touring software using deformable registration algo- such as hypoxia or proliferation, or response pro-
rithms to re-contour a patient’s altered anatomy in a cesses such as cell death, measured at a voxel-level
matter of seconds. Advanced planning tools have using advanced PET or MR imaging; these altera-
already been developed (currently available as a tions can be converted to voxelized prescription
research module only) that can replan an optimized functions, based on prospective evaluation of change
treatment to this volume in just a few minutes. This in function versus local control/relapse rates. An
entire process, completed in a few minutes would initial agnostic radiotherapy prescription can be
allow the assessment of the patient with an MvCT to altered based on patient- and tumor-specific changes
determine whether plan adaption is necessary, per- in these imaging studies, permitting an extremely
form auto-contoured deformable image fusion, sophisticated form of dose painting. Tomotherapy is
recalculate a new plan, and deliver it, all within one of the best radiation treatment modalities suited
minutes. This can therefore allow for frequent, and if for this type of adaptation (Bentzen and Gregoire
necessary, even daily re-planning for certain 2011).
732 D. Khuntia and M. P. Mehta

brain radiotherapy plus stereotactic radiosurgery or radio-


4 Conclusion surgery alone. Int J Radiat Oncol Biol Phys 68(5):
1388–1395
Belderbos JS, Heemsbergen WD et al (2006) Final results of a
In the last decade, advances in stereotactic body Phase I/II dose escalation trial in non-small-cell lung cancer
radiotherapy for medically inoperable lung cancer using three-dimensional conformal radiotherapy. Int J
have yielded local control rates approaching or Radiat Oncol Biol Phys 66(1):126–134
Bentzen SM, Gregoire V (2011) Molecular imaging-based dose
exceeding 90%. Several trials, such as RTOG 0618 painting: a novel paradigm for radiation therapy prescrip-
are now exploring the use of SBRT in medically tion. Semin Radiat Oncol 21(2):101–110
operable stage I and II NSCLC. This approach will Chang EL, Wefel JS et al (2009) Neurocognition in patients
need to become even more sophisticated as we gar- with brain metastases treated with radiosurgery or radio-
surgery plus whole-brain irradiation: a randomised con-
ner a better understanding of the toxicities associated trolled trial. Lancet Oncol 10(11):1037–1044
with it, such as chest wall pain, trachea-vascular Chi A, Jang SY et al (2011) Feasibility of helical tomotherapy
complications, etc., but also because the recent NCI- in stereotactic body radiation therapy for centrally located
sponsored trial of screening high-risk patients with early stage nonsmall-cell lung cancer or lung metastases. Int
J Radiat Oncol Biol Phys (Epub ahead of print, Jan 19)
CT scans has demonstrated survival improvement, Dawson LA, Jaffray DA (2007) Advances in image-guided
thereby ensuring that within a short period of time, radiation therapy. J Clin Oncol Official J Am Soc Clin
CT-screening will become routine. This will lead to Oncol 25(8):938–946
the diagnosis of early stage lung cancer in many DeAngelis LM, Delattre JY et al (1989) Radiation-induced
dementia in patients cured of brain metastases. Neurology
medically inoperable patients, for whom such body 39(6):789–796
stereotactic approaches will become critical. Gondi V, Tolakanahalli R et al (2010a) Hippocampal-sparing
For patients with more locally advanced disease, whole-brain radiotherapy: a ‘‘how-to’’ technique using
dose escalation has resulted in improvement in local helical tomotherapy and linear accelerator-based intensity-
modulated radiotherapy. Int J Radiat Oncol Biol Phys
control as well as survival (Yuan et al. 2007). Fur- 78(4):1244–1252
ther investigations are underway (RTOG 0617) Gondi V, Tome WA et al (2010b) Estimated risk of perihip-
attempting to validate the role of high doses of pocampal disease progression after hippocampal avoidance
radiation with concomitant chemotherapy, and during whole-brain radiotherapy: safety profile for RTOG
0933. Radiother Oncol J Eur Soc Therap Radiol Oncol
advanced technologies such as tomotherapy are 95(3):327–331
ideally suited to precisely and safely deliver highly Hodge W, Tome WA et al (2006) Feasibility report of image
conformal thoracic radiation. guided stereotactic body radiotherapy (IG-SBRT) with
Adaptive radiotherapy will likely be the next tomotherapy for early stage medically inoperable lung
cancer using extreme hypofractionation. Acta Oncol
advance in radiation therapy. Lung cancer patients are 45(7):890–896
ideal candidates for this as their tumors can indeed Khuntia D, Brown P et al (2006) Whole-brain radiotherapy in
change both anatomically and biologically. Techno- the management of brain metastasis. J Clin Oncol Official J
logical advances in computer processing, exit beam Am Soc Clin Oncol 24(8):1295–1304
Kong FM, Ten Haken RK et al (2005) High-dose radiation
dosimetry from MVCTs, and software updates in improved local tumor control and overall survival in
deformable registration will allow for rapid transla- patients with inoperable/unresectable non-small-cell
tion of new plans perhaps on a daily basis to deliver lung cancer: long-term results of a radiation dose
the ultimate adaptive radiation delivery. escalation study. Int J Radiat Oncol Biol Phys 63(2):
324–333
Mackie TR, Balog J et al (1999) Tomotherapy. Semin Radiat
Oncol 9(1):108–117
Mackie TR, Kapatoes J et al (2003) Image guidance for precise
References conformal radiotherapy. Int J Radiat Oncol Biol Phys
56(1):89–105
Mehta M, Scrimger R et al (2001) A new approach to dose
Adkison JB, Khuntia D et al (2008) Dose escalated, hypofrac- escalation in non-small-cell lung cancer. Int J Radiat Oncol
tionated radiotherapy using helical tomotherapy for inoper- Biol Phys 49(1):23–33
able non-small cell lung cancer: preliminary results of a Mizumatsu S, Monje ML et al (2003) Extreme sensitivity of
risk-stratified phase I dose escalation study. Technol Cancer adult neurogenesis to low doses of X-irradiation. Cancer
Res Treat 7(6):441–447 Res 63(14):4021–4027
Aoyama H, Tago M et al (2007) Neurocognitive function of Monje ML, Mizumatsu S et al (2002) Irradiation induces neural
patients with brain metastasis who received either whole precursor-cell dysfunction. Nat Med 8(9):955–962
Advances in Radiation Oncology of Lung Cancer 733

Monje ML, Toda H et al (2003) Inflammatory blockade restores cancer during intensity-modulated radiotherapy: how reli-
adult hippocampal neurogenesis. Science 302(5651):1760–1765 able, consistent, and meaningful is the effect? Int J Radiat
Nagata Y, Takayama K et al (2005) Clinical outcomes of a Oncol Biol Phys 66(1):135–141
phase I/II study of 48 Gy of stereotactic body radiotherapy Song CH, Pyo H et al (2010) Treatment-related pneumonitis
in 4 fractions for primary lung cancer using a stereotactic and acute esophagitis in non-small-cell lung cancer patients
body frame. Int J Radiat Oncol Biol Phys 63(5):1427–1431 treated with chemotherapy and helical tomotherapy. Int J
Perez CA, Stanley K et al (1980) A prospective randomized Radiat Oncol Biol Phys 78(3):651–658
study of various irradiation doses and fractionation sched- Timmerman R, McGarry R et al (2006) Excessive toxicity
ules in the treatment of inoperable non-oat-cell carcinoma when treating central tumors in a phase II study of
of the lung. Preliminary report by the Radiation Therapy stereotactic body radiation therapy for medically inoperable
Oncology Group. Cancer 45(11):2744–2753 early-stage lung cancer. J Clin Oncol Official J Am Soc Clin
Schubert LK, Westerly DC et al (2009) A comprehensive Oncol 24(30):4833–4839
assessment by tumor site of patient setup using daily MVCT Yoon SM, Choi EK et al (2006) Clinical results of stereo-
imaging from more than 3, 800 helical tomotherapy tactic body frame based fractionated radiation therapy for
treatments. Int J Radiat Oncol Biol Phys 73(4):1260–1269 primary or metastatic thoracic tumors. Acta Oncol 45(8):
Shah AP, Langen KM et al (2008) Patient dose from 1108–1114
megavoltage computed tomography imaging. Int J Radiat Yuan S, Sun X et al (2007) A randomized study of involved-field
Oncol Biol Phys 70(5):1579–1587 irradiation versus elective nodal irradiation in combination
Siker ML, Tome WA et al (2006) Tumor volume changes on with concurrent chemotherapy for inoperable stage III
serial imaging with megavoltage CT for non-small-cell lung nonsmall cell lung cancer. Am J Clin Oncol 30(3):239–244
Image-Guided Radiotherapy in Lung Cancer
Percy Lee and Patrick Kupelian

Contents Abstract
Technological advances in image guidance have led
1 Introduction.............................................................. 735 to improvements in outcome for lung cancer patients
2 Image-Guided Target Delineation Using treated with radiation therapy. In particular advance-
Functional Imaging.................................................. 736 ments in 2-Dimensional to now 3 and 4-Dimensional
3 Lung Tumor Motion ............................................... 737 treatment planning as well as the use of functional
imaging such as FDG-PET/CT have allowed for
4 Treatment Room Planar Imaging ......................... 738
more accurate targeting of radiation therapy to areas
5 Fiducial Markers ..................................................... 739 of disease while limiting collateral damage to
6 Treatment Room Volumetric Imaging.................. 740 surrounding normal tissue. Patient specific margins
are now applied to individual patients and are
7 Conclusion ................................................................ 741
substantially less than what has been used in the
References.......................................................................... 741 previous era. This has allowed for radiation dose-
escalation to the tumor while maximally sparing the
surrounding organs at risk and has translated into
improved tumor control and decrease in toxicities
clinically. In-treatment room stereoscopic X-ray
imaging with or without implanted fiducial markers
as well as volumetric kilovoltage or megavoltage
cone-beam CT have allowed clinician to reliably
deliver the planned treatment daily to the intended
target despite the smaller margins of error. Image-
guidance is an important evolving component of
radiation therapy for lung cancer and is an area of
intense research interest.

1 Introduction

P. Lee (&)  P. Kupelian The recent technological advances in radiation ther-


Department of Radiation Oncology, apy have dramatically improved outcomes in lung
University of California at Los Angeles, cancer patients treated with radiation. In the era of
200 UCLA Medical Plaza, Ste. B265,
Los Angeles, CA 90095-6951, USA two-dimensional (2D) radiation therapy, tumor con-
e-mail: percylee@mednet.ucla.edu trol was poor, and dose was limited due to the

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 735
DOI: 10.1007/174_2011_287, Ó Springer-Verlag Berlin Heidelberg 2011
736 P. Lee and P. Kupelian

increased toxicity necessitated by the poor imaging


quality during planning and treatment, and the 2 Image-Guided Target Delineation
requirement to include a large volume of normal tis- Using Functional Imaging
sue in the treatment fields. Three-dimensional (3D)
conformal radiotherapy has incrementally improved In 3D conformal radiotherapy, delineation of the
our outcomes due to better tumor and organ spatial gross tumor volume (GTV) is done primarily with
delineation, but was still unable to account for real- anatomically based CT imaging. Limitations of CT-
time tumor motion. More recently, the use of hypo- based imaging to identify the GTV include variability
fractionated radiation therapy or stereotactic body in window and level settings, motion artifact, indis-
radiation therapy (SBRT) using sophisticated image- tinct tissue boundaries between tumor and lung
guidance has drastically reduced the overall treatment atelectasis that have similar density, and inability to
time as well as improved the tumor control rates to identify pathologically involved lymph nodes less
greater than 90% in medically inoperable stage I non- than 1 cm. By compensating for these inadequacies
small cell lung cancer (NSCLC) (Timmerman et al. using excessive large margins, one adds excessive
2010). This has been possible due to combined treatment toxicities. On the other hand, undercon-
advances in functional and anatomical imaging, touring is likely to lead to inadequate dose to the
improved accuracy and precision of radiation therapy intended target, and local failure due to geographic
delivery devices, advent of four-dimensional (4D) CT miss. For example, using CT-based treatment plan
planning where individualized tumor motion can be often leads to undercontouring of involved lymph
accounted for and characterized, as well as on-board nodes less than 1 cm. One study found that up to 44%
stereoscopic and volumetric imaging in the radiation of nodes involved were less than 1 cm in diameter,
therapy treatment room prior to radiation delivery. and 18% of patients with pathologically involved
The main reasons for local failure after radiation mediastinal lymph nodes did not have any nodes
therapy are (1) inadequate staging imaging to identify larger than 1 cm (Prenzel et al. 2003).
all areas of gross and microscopic disease; (2) geo- Functional imaging such as flurodeoxyglucose
graphic misses due to tumor motion as a result of (FDG)-PET/CT is quite useful for disease staging and
respiration during treatment delivery; and (3) inade- treatment volume delineation for radiation therapy
quate planned radiation dose due to nearby dose- (Bradley et al. 2004; Mac Manus and Hicks 2007).
limiting objects at risk (OARs). Image-guided radiation Most commonly, an FDG-PET/CT can assist in
therapy (IGRT) based on functional planning imaging detecting occult involved mediastinal and hilar lymph
such as positron emission tomography/computed nodes less than 1 cm and distinguish collapsed lung
tomography (PET/CT) as well as 4D CT to account for from tumor (Fig. 1). There are also limitations for
individualized tumor and organ motion, and in-room PET-based image-guided treatment planning. The
image-guidance such as stereoscopic imaging or volu- spatial resolution of PET is relatively poor ([1 cm),
metric cone beam CT (CBCT) has allowed radiation poor temporal resolution due to tumor and organ
oncologist to dose-escalate the radiation to the intended motion as well as non-tumor related nonspecific
target, while reducing dose to OARs, thereby reducing uptake due to inflammation. Specifically for radio-
treatment-related toxicities. A dose–response relation- therapy planning, there are many unanswered ques-
ship for tumor control likely exists for NSCLC as seen tions such as how to best use FDG-PET guided
in the radiation therapy oncology group (RTOG) 7301. imaging for planning. Mainly, several approaches
Trial for stage III NSCLC that established 60 Gy in 30 have been proposed for defining FDG-based GTV: (1)
fractions as the standard dose (Perez et al. 1986). In visual interpretation of the FDG-PET/CT imaging; (2)
medically inoperable stage I NSCLC, with very con- using a percentage such as 40–50% of the maximal
formal SBRT approaches and IGRT, very high bio- uptake as the threshold for target delineation; or (3)
logical doses (up to 20 Gy per fraction) can be delivered using an absolute threshold such as a standard uptake
to peripheral tumors to achieve excellent local control value (SUV) of greater than 2.5 to segment the GTV.
(80–90%) suggesting that local control is possible when Despite its limitations however, the use of FDG-PET/
sufficiently high doses are delivered (Timmerman et al. CT in radiation treatment planning has significantly
2010; Onishi et al. 2004; Xia et al. 2006). improved radiation target delineation, and reduced
Image-Guided Radiotherapy in Lung Cancer 737

geometric uncertainty to the radiation treatment.


In the era of 3D conformal radiotherapy, excessive
large margins were used due to uncertainty regarding
the extent of the tumor motion. However, the devel-
opment of 4D CT with multislice detectors and faster
imaging reconstruction has facilitated the ability to
obtain images while patients breathe and assess tumor
and organ motion (Nehmeh et al. 2004a, b). One
approach to designing individualized target volume
based on the 4D CT is by contouring the lung tumor
in the extreme phases of respiration (end inspiration
and end expiration) during normal breathing. Using
this approach, an internal target volume (ITV) can be
defined. The ITV is defined as the GTV plus an
internal margin (IM) to account for intra and inter-
fractional tumor motion from respiration. Most lung
tumors have a respiratory excursion of less than
5 mm. However, a minority, approximately 10%, of
lung tumors move more than 10 mm and occasionally
motion of 40 mm can be seen, especially in lowerlobe
tumors near the diaphragm (Liu et al. 2007). The use
of an ITV allows patient-specific IM to be applied
Fig. 1 Top axial CT slice of a patient with medically inoperable, rather than population-derived values, the latter can
centrally located, stage I squamous cell carcinoma of the left substantially underestimate or overestimate the IM.
lower lobe causing collapse of the left lower lobe. Bottom PET Tumor motion in the thorax has been assessed in
image of the equivalent axial slice demonstrating FDG avidity of
multiple studies. In one study, the 3D motion of the
only the tumor and not the collapse left lower lobe. PET was used
for stereotactic body radiation therapy image-guided treatment 20 lung tumors was assessed by implanting a 2-mm
planning to target the FDG-avid tumor only gold fiducial marker in or near the tumor. The 3D
position of the implanted markers was determined
geographic misses compared to CT-based treatment using two in-room fluoroscopy imaging processor
planning. Currently, investigation is ongoing to units. The system provided coordinates of the gold
develop other novel PET tracers that are specific for marker during beam-on and beam-off time in all
other biological pathways of cancer, such as hypoxia, directions (Seppenwoolde et al. 2002). On average,
cell proliferation, angiogenesis, apoptosis, and gene lower lobes tumor had the greatest motion in the
expression. These novel functional imaging tools may cranial–caudal dimension (12 ± 2 mm). The lateral
further assist in image-guided radiation therapy and anterior–posterior directions were small both for
planning in the near future. A possibility in the future upper and lowerlobe tumors (2 ± 1 mm). Since the
is to use functional information such as radioresistant tumor spends more time in the exhalation than in the
subvolumes within the GTV and dose-escalated the inhalation phase, the time-averaged tumor position
radiation using intensity-modulated radiation therapy was closer to the exhale position. In another study,
approaches to these subvolumes. using a baseline free-breathing CT scan, the patient’s
couch position was fixed where each tumor showed
its maximum diameter on the image. For 16 tumors,
3 Lung Tumor Motion over 20 sequential CT images were taken every 2 s,
with a 1-s acquisition time occurring during free
Lung tumor motion is a major obstacle in accurate breathing (Shimizu et al. 2000). In the sequential CT
treatment delivery for thoracic malignancies. Respi- scanning, the tumor was not visible in the examina-
ratory-induced target motion (also known as intra- tion slice in 21% (75/357) of cases. There were sta-
fraction tumor motion), can add significant amount of tistically significant differences between lowerlobe
738 P. Lee and P. Kupelian

tumors (39.4%; 71/180) and upperlobe tumors (0%; (De Neve et al. 1992). Typically, the source energy of
0/89; P \ 0.01) and between lowerlobe tumors and the linear accelerator with megavoltage (MV) energy
middle-lobe tumors (8.9%; 4/45; P \ 0.01) in the was used to produce these EPIDs. This produced
incidence of the disappearance of the tumor from the image of poor quality and high imaging dose, while
image. Michalski et al. (2008) evaluated the repro- providing no soft tissue information. Alignment can
ducibility of respiration-induced lung tumor motion be verified using EPIDs either on a daily basis or on
by obtaining 23 pairs of 4D CT scans for 23 patients. another schedule.
The group confirmed that the largest extent of respi- More recently, some of the limitations for onboard
ration-induced motion was along the cranio-caudal megavoltage EPIDs have been overcome by the use
direction, which ranged from \0.5 to 3.59 cm. of kilovoltage (kV) X-ray source to produce kilo-
Furthermore, three patients had dissimilar respiratory- voltage EPIDs. The source is offset at right angles to
induced motion on repeated 4D CT imaging. The the gantry of the linear accelerator head, opposite to a
authors concluded that target motion reproducibility flat-panel detector. Kilovoltage EPIDs produces
was seen in 87% of cases, and advised verification of superior image quality over megavoltage EPIDs while
target motion during treatment. delivering a lower imaging dose to the patient.
Another aspect of tumor motion relates to its Orthogonally mounted EPIDs in room can be used
potential for deformation during the respiration cycle. to track bone and fiducial markers in real time
Wu et al. (2009) evaluated the extent of tumor (Seppenwoolde et al. 2002; Shirato et al. 2000). The
deformation, along with motion in the 3D for 30 Cyberknife system (Accuray, Sunnyvale, CA) mounts
patients with early stage NSCLC. They evaluated the a 6-MV miniature linear accelerator source on a
overlap index after accounting for translation of only robotic arm and is equipped with an in-room
image registration, after accounting for translation orthogonal EPIDs system. The robotic arm is able to
and rotation, and also after accounting for transla- move and track a moving lung tumor by tracking
tions, rotation, and deformation. The overlap index either implanted fiducial markers or soft-tissue cor-
increased only by 1.1 and 1.4%, respectively, when relate, and make adjustments to the beam. Similarly,
one accounted for rotation and rotation plus defor- the ExacTrac/Novalis Body System (BrainLAB AG,
mation. These results were independent of GTV size Heimstetten, Germany) has flat-panel detectors built
and motion amplitude. The authors concluded that the in the treatment room that detects kilovoltage sources
primary effect of normal respiration on tumor motion from the floor. Treatment adjustments and tracking
was the translations of tumors and while tumor rota- can be performed when internal surrogate markers are
tion and deformation played a minimal role. used (Fig. 2). These stereoscopic IGRT approaches
are particularly useful for treatment of tumors in the
central nervous system, or spine applications as bone
4 Treatment Room Planar Imaging is readily detected with these images in realtime and
are stable surrogate markers. One advantage of planar
Most of the beneficial effects of IGRT are seen in the IGRT over volumetric-based IGRT such as CBCT is
treatment room. As previously mentioned, due to the speed at which images are acquired and adjust-
setup uncertainty and lack of image-guidance, large ments can be made. For both Accuray and BrainLab
planning margins were traditionally added to ensure systems, an automated 2D/3D co-registration algo-
that the target is encompassed in the PTV due to setup rithm is applied to align a 3D CT patient data set with
uncertainty and tumor motion. This can lead to a 2 kV-images. When properly calibrated such that the
substantial amount of unnecessary irradiation to nor- exact position of the kilovoltage tubes and detectors
mal tissue causing excessive toxicities, or limiting the are known with respect to the machine’s isocenter, it
dose to the tumor that led to local recurrence. For is possible to generate DRRs from planning CT at the
example, adding a 0.5-cm thick shell to a spherical same positions as the kilovoltage images are acquired
GTV of 4.5 cm in diameter nearly doubles its volume. and compare with the acquire kilovoltage images. An
Traditionally, electronic portal imaging devices automated registration algorithm based on gradient
(EPIDs) acquiring 2D orthogonal views were used to correlation is used, and corresponding shifts in 3D are
align and verify treatment position and isocenter derived.
Image-Guided Radiotherapy in Lung Cancer 739

Fig. 2 Top DRR to


kilovoltage matching using
spine anatomy using paired
oblique stereoscopic imaging
in the treatment room. Bottom
DRR to kilovoltage matching
using implanted fiducial
markers in the left upper lobe
lung tumor during image-
guided SBRT

such as pneumothorax, bleeding, or infection.


5 Fiducial Markers A secondary disadvantage is the delay in the therapy
in order for the markers to be placed and stabilized in
For these planar IGRT systems to be able to gate or the patient’s body. For the thorax, marker implanta-
track the tumor during radiation treatment, radi- tion can be done either transcutaneously or trans-
opaque markers need to be implanted in or near the bronchially. One study reported marker implantation
intended target. The fiducial markers act as internal in 15 patients performed transcutaneously and eight
radiologic landmarks and are assumed to move with a patients transbronchially (Kupelian et al. 2007). Eight
constant relationship to the targeted tumor during of the 15 transcutaneous implants developed pnemo-
therapy. The main disadvantage of using a marker- thoraces, six of which required the placement of a
based system for IGRT is the risk of complications chest tube. None of the patients who underwent
740 P. Lee and P. Kupelian

transbronchial implantation developed pneumothorax. superior–inferior direction for intra- and inter-frac-
The authors concluded that in their patient population tional variations. Tumor visibility was sufficient for
with many elderly patients with emphysema, trans- markerless tracking in 47% of the EPID movies.
cutaneous marker implantation led to a high compli- Another study compared two respiratory surrogates for
cation rate compared to the transbronchial approach. gated lung radiotherapy without internal fiducials
Kothary et al. (2009) reported a similar study of (Korreman et al. 2006). Video clips were acquired after
percutaneous fiducial implantation in 132 patients six patients had fiducial markers implanted in lung
(139 implants) . Of the 139 implantations, 44 were in tumors to be used for image-guided SBRT. The posi-
the lung. Pneumothoraces were seen in 20 of 44 lung tions of the markers in the clips were measured within
implantations (45%); a chest tube was required in the video frames and used as the standard for tumor
only seven of the 44 lung implantations (16%). Of the volume motion. Two external surrogates, a fluoro-
139 implantations, 133 were successful with six scopic image correlation surrogate and an external
procedures leading to marker migration (4.3%). The optical surrogate were compared to the standard. In
authors concluded that percutaneous fiducial implan- four out of the six cases, fluoroscopic image correlation
tation is safe and effective with acceptable risks surrogate was superior to the external optical surrogate
similar to conventional percutaneous organ biopsy. in the AP-views. In one of the remaining two cases, the
Thus, it would appear that the risk of complications two surrogates performed comparably. In the last case,
from percutaneous marker implantation is high the external fiducial surrogate performed best. The
dependent on the patient population as well as the authors concluded that fluoroscopic gating based on
experience of the operator. Nevertheless, implanting correlations of native image features may be adequate
fiducial markers require extra effort and time, and for respiratory gating.
delays initiation of treatment.
Overall, stereoscopic kilovoltage-imaging has some
distinct advantages and disadvantages. Volumetric 6 Treatment Room Volumetric
data assessment is difficult based on the planar images. Imaging
The system will always require an internal surrogate to
identify the target, track, or gate the treatment, be it More recently, the implementation of in-room volumet-
bony anatomy, or radiopaque markers. Complications ric imaging has extended the possibilities of image
from marker implantation and marker migration after guidance. The availability of volumetric imaging with
implantations are always valid concerns. The limita- kilovoltage or megavoltage helical CT or cone-beam CT
tions of the planar imaging, however, are compensated (CBCT) scans has allowed the evolution of image guid-
by the simultaneous stereoscopic image acquisitions ance for lung cancers in three fundamental ways: (1) as an
allowing for six degrees of freedom positioning alternative method for guidance not relying on implanted
assessment, especially when combined with a robotic fiducials, (2) assessment of anatomic variations in soft
couch system (Novalis) or robotic arm (Cyberknife), tissues during a treatment course (e.g. relative location of
which makes the process efficient and faster than tumor and relevant organs at risk, changes in tumor size),
obtaining volumetric imaging. Also, this system oper- and (3) the future possibility of assessing dosimetric
ates independent of the treatment allowing image variations in radiation delivery either on a daily basis or
acquisition during treatment, and real-time assessment cumulatively throughout a treatment course.
of target motion in order to compensate for intrafrac- For tumor localization, in-room CT scans are ade-
tional tumor and organ motion. quate to localize lung tumors even though images are
Due to the limitations of a marker-based system in typically inferior in quality compared to multi-slice
planar-based IGRT, groups have evaluated the feasi- kilovoltage helical CT scans. In addition, these are
bility of using respiratory surrogates without internal essentially ‘‘slow’’ scans that will give an ‘‘average’’
fiducials. One study evaluated the feasibility for position of a mobile tumor. Motion ‘‘tracking’’ is not
markerless tracking of lung tumors in SBRT (Richter easily implemented at this stage using in-room CT
et al. 2010). EPID movies were acquired during SBRT scans. However, the fact that the scans provide the
treatment given to 40 patients with 49 lung targets, and ‘‘average’’ position of a mobile lung tumor makes such
retrospectively analyzed via 4D CT and EPID in the scans adequate for initial positioning of patients on a
Image-Guided Radiotherapy in Lung Cancer 741

Fig. 3 In-room megavoltage CT scan of the chest demonstrating changing tumor size throughout a treatment course

regular basis throughout a treatment course, eliminat- respect to being utilized for dose evaluations.
ing both systematic and random errors in initial set ups. However, they do enable assessment of dosimetric
Even if gating is not performed on the basis of breathing implications of shrinking tumor, changing shapes, and
cycle signals, a motion envelope can be safely designed deforming organs.
around this ‘‘average’’ tumor position. Implanted
fiducials and the associated implantation potential
toxicities are therefore avoided. This avoidance of 7 Conclusion
additional procedures, such as trans-cutaneous or trans-
bronchial metallic fiducial implanation, in this group of Image guidance in the treatment of lung cancer is
patients with typically poor general health is an performed with a variety of techniques ensuring the
important advantage of volumetric imaging. accuracy of radiation delivery. These techniques are
In-room volumetric imaging also allows for a essential to major advances in lung cancer treatments
better evaluation of anatomic variations throughout a such as stereotactic body radiotherapy delivering
course of treatment beyond the simple location of an ablative doses. It is important to realize that these
intrathoracic tumor (Fig. 3). Information about tumor techniques are still works in progress. Future appli-
progression or response, relative position of primary cations utilizing in-room images are being developed,
lesions versus lymph node changes that could be such as adaptive therapy. Overall, image guidance has
affected by deformations or rotations, development or resulted in significant refinement of radiation delivery
resolution of atelectatic areas can be obtained. These techniques and is an integral part of future advances in
are situations that cannot be assessed without volu- lung cancer radiotherapy.
metric imaging. One such important future applica-
tion of tumor response assessment in patients with
locally advanced lung cancer is the ability to identify
References
individual patients who have significant tumor
response at some point throughout treatment and Bradley J, Thorstad WL, Mutic S et al (2004) Impact of FDG-
PET on radiation therapy volume delineation in non-small-
subsequently might benefit from implementation of cell lung cancer. Int J Radiat Oncol Biol Phys 59:78–86
adaptive therapy approaches. Such approaches will De Neve W, Van den Heuvel F, De Beukeleer M et al (1992)
push dose escalation beyond currently used doses. Routine clinical on-line portal imaging followed by imme-
Finally, the availability of soft tissue images does diate field adjustment using a tele-controlled patient couch.
Radiother Oncol 24:45–54
facilitate potential dosimetric evaluations of delivered Korreman S, Mostafavi H, Le QT et al (2006) Comparison of
therapies. Megavoltage CT scans versus kilovoltage respiratory surrogates for gated lung radiotherapy without
conebeam CT scans present different challenges with internal fiducials. Acta Oncol 45:935–942
742 P. Lee and P. Kupelian

Kothary N, Heit JJ, Louie JD et al (2009) Safety and efficacy of Prenzel KL, Monig SP, Sinning JM et al (2003) Lymph node
percutaneous fiducial marker implantation for image-guided size and metastatic infiltration in non-small cell lung cancer.
radiation therapy. J Vasc Interv Radiol 20:235–239 Chest 123:463–467
Kupelian PA, Forbes A, Willoughby TR et al (2007) Implan- Richter A, Wilbert J, Baier K et al (2010) Feasibility study
tation and stability of metallic fiducials within pulmonary for markerless tracking of lung tumors in stereotactic
lesions. Int J Radiat Oncol Biol Phys 69:777–785 body radiotherapy. Int J Radiat Oncol Biol Phys 78:
Liu HH, Balter P, Tutt T et al (2007) Assessing respiration- 618–627
induced tumor motion and internal target volume using Seppenwoolde Y, Shirato H, Kitamura K et al (2002) Precise
four-dimensional computed tomography for radiotherapy of and real-time measurement of 3D tumor motion in lung due
lung cancer. Int J Radiat Oncol Biol Phys 68:531–540 to breathing and heartbeat, measured during radiotherapy.
Mac Manus MP, Hicks RJ (2007) Impact of PET on radiation Int J Radiat Oncol Biol Phys 53:822–834
therapy planning in lung cancer. Radiol Clin North Am Shimizu S, Shirato H, Kagei K et al (2000) Impact of
45:627–638 respiratory movement on the computed tomographic images
Michalski D, Sontag M, Li F et al (2008) Four-dimensional of small lung tumors in three-dimensional (3D) radiother-
computed tomography-based interfractional reproducibility apy. Int J Radiat Oncol Biol Phys 46:1127–1133
study of lung tumor intrafractional motion. Int J Radiat Shirato H, Shimizu S, Kunieda T et al (2000) Physical aspects
Oncol Biol Phys 71:714–724 of a real-time tumor-tracking system for gated radiotherapy.
Nehmeh SA, Erdi YE, Pan T et al (2004a) Four-dimensional (4D) Int J Radiat Oncol Biol Phys 48:1187–1195
PET/CT imaging of the thorax. Med Phys 31:3179–3186 Timmerman R, Paulus R, Galvin J et al (2010) Stereotactic
Nehmeh SA, Erdi YE, Pan T et al (2004) Quantitation of body radiation therapy for inoperable early stage lung
respiratory motion during 4D-PET/CT acquisition. Med cancer. JAMA 303:1070–1076
Phys 31:1333–1338 Wu J, Lei P, Shekhar R et al (2009) Do tumors in the
Onishi H, Araki T, Shirato H et al (2004) Stereotactic lung deform during normal respiration? An image regis-
hypofractionated high-dose irradiation for stage I nonsmall tration investigation. Int J Radiat Oncol Biol Phys 75:
cell lung carcinoma: clinical outcomes in 245 subjects in a 268–275
Japanese multiinstitutional study. Cancer 101:1623–1631 Xia T, Li H, Sun Q et al (2006) Promising clinical outcome of
Perez CA, Bauer M, Edelstein S et al (1986) Impact of tumor stereotactic body radiation therapy for patients with inop-
control on survival in carcinoma of the lung treated with erable Stage I/II non-small-cell lung cancer. Int J Radiat
irradiation. Int J Radiat Oncol Biol Phys 12:539–547 Oncol Biol Phys 66:117–125
Proton Therapy for Lung Cancer:
State of the Science
David A. Bush

Contents Abstract
Proton radiation for cancer offers to the physician
1 The Clinical Problem .............................................. 744 the ability to conform the high-dose region to
2 Physical Characteristics of Proton Radiation the tumor while reducing the dose of radiation to
Therapy..................................................................... 744 adjacent normal tissues. In lung cancer this results
3 Treatment-Planning Considerations in the ability to deliver higher doses to the targeted
and Comparisons ..................................................... 746 volume while yet permitting greater sparing of
uninvolved and critical normal tissues: lung, heart,
4 Considerations in Delivering Proton Beams
for Lung Cancer ...................................................... 748 esophagus, and spinal cord. Studies comparing the
distribution of radiation dose show that proton
5 Clinical Outcomes.................................................... 748
radiation is superior to photon radiation for lung
6 Conclusion ................................................................ 750 cancer, even when factors such as respiratory
References.......................................................................... 750 motion are considered. Clinical experience con-
firms the feasibility of proton radiation for early-
stage non-small-cell lung cancers; clinical trials
are being performed in locally advanced tumors.
Evidence accumulated thus far indicates that
proton radiation should be further explored.

Abbreviations

BID Twice-daily treatment


CGE (sometimes expressed as GyE) Cobalt gray
equivalent, a means of expressing the
proton dose in relation to Gray units
(cf below) while allowing for the slightly
higher radiobiologic effect of protons
(i.e., 1.1 in relation to cobalt)
D. A. Bush (&) CTV Clinical target volume
Department of Radiation Medicine, GTV Gross tumor volume
Loma Linda University Medical Center, Gy Gray, standard radiation dose. A common
11234 Anderson Street, Loma Linda,
California 92354, USA daily fraction of photon-beam radiation
e-mail: dbush@dominion.llumc.edu is 2 Gy (or 200 centigray (cGy)); a

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 743
DOI: 10.1007/174_2011_256, Ó Springer-Verlag Berlin Heidelberg 2011
744 D. A. Bush

30-fraction course of radiation at that dose tissue regions such as the heart, lung, esophagus, and
rate equates to a total dose of 60 Gy spinal cord. Tumor-control rates with photon radia-
IMPT Intensity-modulated proton therapy tion therapy, however, continue to be disappointing,
IMRT Intensity-modulated radiation therapy in part because of dose-limiting constraints associated
NSCLC Non-small-cell lung cancer with these normal structures.
PTV Planned target volume
SBRT Stereotactic body radiation therapy

2 Physical Characteristics of Proton


Radiation Therapy

1 The Clinical Problem Because protons (hydrogen ions) have mass (1835
times that of an electron) and an electric charge, the
Although death rates have declined in men and physician can control the beam in three dimensions
recently have stabilized in women, presumably due to and thus can deposit radiation doses more accurately
reductions in cigarette smoking, lung cancer remains within target volumes while minimizing—or, often,
one of the leading causes of cancer death in North eliminating—the radiation dose to surrounding non-
America (Jemal et al. 2010). Non-small-cell lung targeted tissues. Mass reduces the extent to which the
cancers (NSCLC) predominate over the small-cell particle scatters as it passes through tissue; charge
variant and are usually associated with a poor prog- enables interaction with orbiting electrons within
nosis because most patients have locally advanced or targeted cells, and hence the ionization required to
metastatic disease when first seen. The only NSCLC begin the process of cell destruction; and beam energy
subgroup associated with a better than 50% 5-year enables the radiation oncologist to control depth of
survival rate consists of individuals having peripher- penetration. The ability to spare normal tissues is
ally located T1 or T2 tumors without evidence of important. The greater the extent to which the phy-
nodal or distant metastases (Mountain 1997). More sician can reduce or eliminate the dose to normal
than 80% of patients, however, present with more- tissues, the lesser is the likelihood that treatment may
advanced stages of disease. need to be compromised or halted because of unac-
The goal of definitive radiotherapy is to eradicate ceptable side effects. The ability to reduce or elimi-
intra-thoracic disease while respecting the radiation nate radiation dose to normal tissues, thanks to the
tolerance of nearby normal structures by minimizing reduced lateral scatter and sharp dose fall-off of the
the dose to such structures. Various photon (X-ray) proton-beam, not only allows one to deliver the total
techniques have been employed to effect a therapeutic needed dose but also yields opportunities to deliver
advantage, among them are hyperfractionation (mul- higher doses without increasing side effects.
tiple treatments per day); accelerated fractionation The importance of reducing the volume integral
(shorter treatment periods); and dose escalation dose to normal tissues has long been noted. In studies
(Byhardt et al. 1993; Kong et al. 2005; Rosenman spanning more than 40 years, Philip Rubin and sev-
et al. 2002; Saunders et al. 1997). Most innovative eral collaborators identified the clinicopathologic
techniques have focused on conformal treatment courses of radiation injury in organs and tissues
delivery with computer-assisted three-dimensional throughout the body, and also identified tolerance
therapy planning and, in some cases, intensity-mod- doses for those organs. Tolerances were identified in
ulated photon radiotherapy (IMRT) or stereotactic ranges of total doses in which severe or life-threat-
body radiation therapy (SBRT), in which more-com- ening complications were likely to occur within five
plex treatment-planning and delivery can allow the years of therapeutic radiation; i.e., severe sequelae
radiation oncologist to have better control of doses to would likely occur in 5% of patients treated at the
healthy tissues (Grills et al. 2003; Murshed et al. lower end of the range (TD5/5) and in 50% of patients
2004). The intent has been to deliver higher doses to treated to the dose at the top of the range (TD50/5)
target volumes in an effort to improve local tumor- (Rubin et al. 1978). Although organs and tissues were
control within the constraints of surrounding normal- separated into categories according to their
Proton Therapy for Lung Cancer: State of the Science 745

Fig. 1 Percent dose deposited per depth in tissue for photon


beams of various energies, and a proton-beam (shown in red)

importance for survival, no ‘‘safe’’ dose (TD0/5) was


identified for any organ; rather, in a classic series of
graphs, Rubin and Casarett (1968) demonstrated that
sublethal doses of radiation initiate a course that can
eventuate in clinical manifestations of radiation
injury, some of which progress to lethality. Studies
such as these highlight the desirability of avoiding
radiation exposure to normal tissues whenever pos-
sible, yet paradoxically, the present ‘‘gold standard’’
of photon radiotherapy, IMRT, increases the volume
of tissues exposed to low doses.
In later studies, Rubin et al. demonstrated early
and persistent elevation of cytokine production fol-
lowing pulmonary irradiation. The temporal rela-
tionship between elevation of specific cytokines and
histological and biochemical evidence of fibrosis
illustrated the continuum of response which, the
Fig. 2 Planning comparison of a single lateral field with a X-
authors speculated, underlies pulmonary radiation ray (left) and proton-beam (right)
reactions and supports the concept of a perpetual
cascade of cytokines, produced immediately after
radiotherapy and persisting until expression of path- treatment of similar target volumes with photon
ologic and clinical late effects (Rubin et al. 1995). beams, these properties permit sparing the treated
Proton-beam therapy offers a means to reduce the lung, opposite lung, heart, esophagus, and spinal cord,
volume integral dose. Protons demonstrate a Bragg in turn allowing for safe dose escalation.
peak: the deposited dose is relatively low upon Because local tumor-control rates are generally
entrance and increases slowly until reaching the pre- poor with conventional photon radiation treatment,
scribed depth in tissue; there, the bulk of the dose is and because evidence exists to indicate that higher
deposited within the targeted volume and no dose is doses may improve tumor-control, investigators have
deposited in distal tissues. The radiation oncologist anticipated a role for proton radiation therapy in
can spread out the Bragg peak to encompass the target patients with lung cancer. Fowler (2003), using bio-
volume while still retaining a relatively low entrance logical modeling, estimated that significant improve-
dose and sparing tissues distal to the direction of ments in local tumor-control and survival are likely to
travel (Fig. 1). In NSCLC, and in contrast to supervene with proton radiotherapy. Because of the
746 D. A. Bush

normal-tissue-sparing properties of proton beams, not parameters are greater, respiratory monitoring with
only does an opportunity exist to achieve effective beam gating, or respiratory control or compensation,
disease control without increased normal-tissue perhaps via a patient positioning device that moves
complications, it also may be possible to decrease the the patient during treatment in a way that allows the
severity of toxicity that is seen in comparable treat- target to remain still relative to the treatment beam,
ment regimens with photons. Accordingly, clinical may be employed to significantly reduce the dose to
investigations of proton and other heavy-charged- normal lung tissue.
particle treatments for lung cancer are being con- Reports by Engelsman and colleagues (Engelsman
ducted around the world. and Kooy 2005; Engelsman et al. 2006) compared
computerized treatment plans using various tech-
niques to account for respiratory motion in planning
proton treatments. Target expansion utilizing treat-
3 Treatment-Planning ment planning in which the planning CT data set
Considerations and Comparisons includes multiple phases of the respiratory cycle (the
fourth dimension of 4D planning) was found to pro-
As noted, proton beams exhibit a Bragg peak, which vide the most reliable target coverage. Intrinsic pul-
yields normal-tissue sparing not possible with photon monary or treatment-related factors, such as pleural
beams. This capability persists despite special dosi- effusion or atelectasis, may also cause uncertainties in
metric circumstances for treating lung cancer with the stopping region of the Bragg peak, and thus need
protons; these circumstances differ from those asso- to be accounted for in therapy planning. If the radi-
ciated with proton therapy elsewhere in the body. For ation oncologist suspects that such changes may occur
example, lung cancers are invariably surrounded by during the treatment course, she or he may find it
aerated pulmonary parenchyma, in which an essen- necessary to repeat chest imaging and create a new
tially water-density-equivalent tumor is surrounded therapy plan for any significant anatomic changes.
by substantially less-dense lung tissue. Aerated lung This practice is referred to as adaptive treatment
tissue possesses reduced stopping power compared to planning.
other soft tissues, thus affecting the stopping distance Using four-dimensional computed tomography,
or distal edge of a spread-out Bragg peak; this can Zhao et al. (2007) investigated dosimetric errors caused
cause proton beams to travel some distance beyond by lung tumor motion in order to find an optimal method
the distal edge of the intended target volume, a situ- of designing patient compensators and apertures for a
ation that is not typically seen in tumors located in passive-scattering beam delivery system and treating
other parts of the body. Additionally, when lung tar- the patient under free breathing conditions. In their
gets are selected for treatment, the physician needs to study, the maximum intensity projection method was
consider physiologic internal target motion, which compared to patient-specific internal margin designs
depends greatly on the region of the lung in which the based on a single breathing phase at the end of inhale or
tumor is located: tumors located near the diaphragm, middle of exhale. They found that maximum intensity
for example, will show the largest extent of move- projection provided superior tumor dose distribution
ment owing to respiration. The simplest method compared to patient-specific internal margin designs.
to account for such motion is to measure the Clearly, great care must be taken in planning
three-dimensional excursion of the tumor and expand proton therapy for lung cancer. This care is accentu-
the treatment volume to encompass all possible tumor ated by the proton beam’s energy-deposition sensi-
positions. For small tumors, with motion parameters tivity to variations in density caused by respiratory
of less than 1 to 2 cm, this method is adequate: the motion. Even allowing for such factors, however,
technique has been used for patients with peripheral proton-beam dosimetry improves upon that obtain-
lung tumors of 1 to 4 cm in size at Loma Linda able with photon beams (Fig. 2). The tissue-sparing
University Medical Center (LLUMC) for more than a capability of the proton-beam is most apparent in the
decade, with an excellent safety profile and a low treatment of early NSCLC (Fig. 3), but significant
incidence of radiation pneumonitis (Bonnet et al. sparing also may be achieved when using protons to
2001). For larger tumors, and when motion treat locally advanced disease.
Proton Therapy for Lung Cancer: State of the Science 747

Fig. 3 Dose distribution (Top) for a stage I lung cancer treated percentage of the total prescribed dose. Dose–volume histo-
with proton-beam shown in axial (left) and sagittal views gram (Bottom) for the same plan. Note the great disparity on
(right). Beams include a right lateral, right posterior oblique dose delivered to the CTV in comparison to that received by the
and posterior arrangement. Colored contours represent the spinal cord, heart, esophagus, and lung

Chang et al. (2006) have published a formal dose plans. Doses to the heart, esophagus, and spinal
comparison between proton and photon treatment cord were also found to be significantly reduced. This
planning. They analyzed ten patients with inoperable study suggests that protons can achieve higher total
Stage I lung cancer, comparing proton treatment plans doses to the target volumes, with more significant
to three-dimensional (3D) conformal photon plans at normal-tissue sparing, than 3D conformal radiation
two total dose levels (66 and 87.5 Gy). Analysis therapy or IMRT.
revealed an approximately 50% reduction in non- Another planning comparison indicates that protons
target lung dose when protons where used. The nor- offer a therapeutic advantage in more-challenging
mal lung tissue-sparing effect seemed to be increased Stage I cases as well. Register et al. (2010) evaluated 15
with the high-dose plans, indicating an additional patients with centrally or superiorly located (within
benefit for dose escalation when proton beams are 2 cm of critical structures) SBRT (50 Gy in 4
used. Fifteen patients with Stage III lung cancer were fractions). They compared the plans used for those
also analyzed, comparing 3D photon, IMRT, and patients with treatment plans for passive-scattering
proton-beam plans at two total dose levels (63 and proton therapy and intensity-modulated proton therapy
74 Gy). Again results showed a substantial reduction (IMPT). Both forms of proton plans, but notably the
in the non-target lung dose, and again, the proton IMPT variant, offered superior sparing of critical nor-
benefit seemed to be more evident with the higher- mal structures in these presentations.
748 D. A. Bush

Similarly, a study by many of the same investi- lateral, posterior, and posterior oblique beams. Fre-
gators (Zhang et al. 2010) compared dose–volume quently the lateral beam is preferred, as it typically
histograms (DVHs) for patients with extensive Stage will provide the lowest volume of normal lung tissue
IIIB NSCLC treated with IMRT, passively scattered exposed. This is in distinct contrast to photon beams,
protons beams, and IMPT. The latter spared more which will continue through the mediastinum into the
lung, heart, spinal cord, and esophageal tissue, even contralateral lung. Lateral proton beams, however, are
with dose escalation. Compared with passively scat- very useful for lung cancer treatment, as the Bragg
tered protons, IMPT allowed further dose escalation, peak allows the beams to stop without irradiating tis-
from 74 Gy to a mean maximal tumor dose of sues distal to the tumor; this location frequently is at
84.4 Gy while all parameters of normal-tissue sparing the mediastinal pleural surface, thus completely
were kept at lower or similar levels. They concluded sparing the mediastinum and contralateral lung. Mul-
that IMPT offered opportunities to pursue individu- tiple treatment beams per day have been utilized;
alized radical radiotherapy for such patients. hypofractionated treatment courses have been
common.
Treatment techniques in patients with locally
advanced lung cancer can be significantly more
4 Considerations in Delivering challenging. Mediastinal lymph nodes usually are
Proton Beams for Lung Cancer targeted; then, other sensitive normal-tissue structures
come into play, such as the heart, esophagus, and
Today, most heavy-charged-particle-beam treatment spinal cord. Such patients are being treated with
facilities utilize a beam scattering system and passive protons and chemotherapy at LLUMC under a clinical
beam-shaping devices, such as an aperture to shape trial. Beam arrangements typically used for such
the perimeter of the particle beam and a tissue com- patients include anterior beams, which stop short of
pensator to shape the distal edge or Bragg peak region the spinal cord, along with lateral and posterior
to be in contour with the distal edge of the target. oblique beams. When beams are designed to limit the
Such devices are designed carefully, to avoid unnec- dose to the esophagus or spinal cord, it is generally
essarily over-radiating pulmonary tissue while preferred that the edge of the aperture be used to
allowing for factors such as altered stopping power in protect these structures. If a beam is designed to stop
aerated tissue and physiologic motion (Moyers et al. short of a critical normal-tissue region, great care is
2001). Scanning-beam technology, essential to IMPT, taken to account for physiologic motion and incon-
is under development at several treatment centers; sistent tissue densities within the chest.
most authorities believe this will provide enhanced
target coverage and normal-tissue protection not
achievable with passive beam-shaping methods
commonly used at present. However, the application 5 Clinical Outcomes
of this technology for treating intra-thoracic targets
presents a significant challenge owing to physiologic The proton-beam has been the most commonly used
internal motion and potentially unreliable radiologic particle beam for treating patients with lung tumors.
path lengths. Until there is a thorough understanding The largest reported experience has been with early-
and reliable control of these variables, it is likely that stage NSCLC at LLUMC: patients with clinical Stage
scattered beams will continue to be utilized for lung I disease, who are either medically inoperable or
cancer treatment. refuse a recommended surgical intervention, are
No standardized treatment techniques or beam treated on a clinical trial. The latest report includes 68
arrangements exist for using heavy-charged-particle patients treated with either 51 CGE or 60 CGE in ten
beams in patients with lung cancer. The largest fractions over a two-week course (Bush et al. 2004).
experience is from LLUMC, where proton beams have The area targeted for treatment includes the gross
been employed since 1995. For patients with solitary tumor volume (GTV) as well as a planned target
pulmonary nodules, the beam arrangements employed volume (PTV) that includes additional margin for
have been relatively simple, typically consisting of respiratory motion. There was no size constraint, and
Proton Therapy for Lung Cancer: State of the Science 749

Fig. 5 Dose–volume histogram for the total of the irradiated


lung associated with the treatment plan shown in Fig. 4
Fig. 4 Dose distribution for a patient with stage III lung
cancer treated with proton-beam. Contours represent the lung toxicity, with the majority of these being seen in
tumor and involved nodes (red) and sub-clinically involved
patients with larger tumors. Shioyama et al. (2003)
mediastinal nodes (light blue). Distribution shows the percent
of the total dose. The beam arrangement includes a left lateral have reported on 28 patients with Stage I non-small-
and posterior to the CTV (46 CGE) and a left posterior oblique cell lung cancer treated with proton beams to a
to the GTV (30 CGE) median dose of 76 Gy at 3 Gy per fraction. Patients
with T1 tumors had a 70% overall survival rate at
centrally located tumors were included; SBRT series 5 years, while patients with T2 tumors had a signifi-
usually exclude the latter. Typically, two to four cantly lower survival rate (approximately 16%). Pul-
beams are utilized for treatment, with at least two monary toxicity was reported as minimal. A recent
fields being treated each day. Various beam weigh- report from Nakayama et al. (2010) reaffirms that, in
tings have been employed, generally with preference their experience, proton radiation therapy was effec-
given to lateral beams to minimize lung exposure tive and well tolerated in medically inoperable
(Fig. 3). In this series the median lung v20 was 7% patients with Stage I NSCLC.
and the median v60 was 2%. The therapy has been Studies now underway at LLUMC utilize proton-
exceptionally well tolerated, with a low incidence beam radiotherapy in conjunction with chemotherapy
of grade 1 pneumonitis and no reported grade 3 for treating locally advanced lung cancers. Neo-adju-
toxicities. vant and concurrent chemotherapy is administered with
Disease-specific survival at three years was 73%. proton therapy, which is given as a concomitant boost.
Patients with T1 tumors have achieved local control The dose to the sub-clinically involved mediastinum is
in 87% of cases; those with tumors larger than 3 cm 46 CGE in 2 CGE fractions with a GTV, BID boost of
(T2) have had local failures up to 50% at three years, 30 CGE during the last three weeks of treatment.
which has led to a third dose escalation: the current It should be noted that the mediastinal CTV is
regimen delivers 70 CGE in 10 equally divided omitted from most IMRT trials, owing to concerns for
fractions over two weeks. Although no survival or pulmonary toxicity. Despite the much larger target
local control data are yet available at this dose level, volumes and dose escalation in such cases, significant
there does not appear to be any difference in tolerance sparing of normal pulmonary tissues still is achieved
after treating nearly 40 patients at this escalated total (Figs. 4 and 5). In the most recent evaluation of
dose. No decline of post-treatment pulmonary func- ongoing results in 19 patients, no Grade 3 or 4 esoph-
tion (FEV1 or PO2) has been observed (Bonnet et al. ageal toxicities were observed; Grade 3 leukopenia was
2001). seen in two patients; Grade 3 thrombocytopenia
In Japan, Nihei et al. (2006) reported on 37 occurred in one individual.
patients with Stage I non-small-cell lung cancer Two Phase II clinical trials are currently underway,
treated with protons. Most patients received between testing the use of proton radiotherapy in combination
80 and 88 Gy equivalent, utilizing fraction sizes with concurrent chemotherapy for locally advanced
ranging from 3.5 to 4.9 Gy. The reported two-year (Stage III), inoperable NSCLC. Studies at the
local–regional relapse-free survival rate for T1 tumors University of Florida (ClinicalTrials.gov identifier:
was 79%; for T2 tumors the rate was 60%. They NCT00881712) and the University of Texas M.D.
identified six cases of grade 2 and 3 pulmonary Anderson Cancer Center (ClinicalTrials.gov
750 D. A. Bush

identifier: NCT00495170) are evaluating proton delivery to the intended target and/or decreases the
radiation in combination with Paclitaxel and Carbo- dose to surrounding healthy tissues has been sufficient
platin. These safety/efficacy studies are similar, yet evidence for many. The proton-beam would seem to
differ in some details. In the Florida study, the pri- meet these criteria today. Evidence and experience
mary outcome measure is to determine whether accumulated thus far, therefore, warrant the continued
reduced acute toxicity from combined concomitant exploration of proton therapy for lung cancer. It is
chemotherapy and radiotherapy occurs, compared to highly likely that the modality has not yet reached its
previous cooperative group trials; disease control, full potential in the treatment of such tumors. The
median overall survival, and 5-year survival are next advance likely will be in the routine clinical use
identified as secondary outcome measures. In the of IMPT. As has been noted above, comparisons of
M.D. Anderson study, median survival time is the IMPT plans with scattered-beam proton plans and
primary outcome measure, with local control, pro- IMRT plans show a therapeutic advantage for the
gression-free survival, disease-specific survival, and former. Clinical application of IMPT will require
disease-free survival as secondary outcomes. That reliable control of the variables associated with
study also seeks to determine whether grade 3 and physiologic internal motion and potentially unreliable
higher toxicities are reduced; whether pre- and post- radiologic path lengths, but it is highly likely that
treatment PET/CT are useful in predicting clinical such control will be achieved as has occurred with
outcome; and whether a biomarker can be used for IMRT. At that point, IMPT may become an effective
predicting treatment response and toxicities. Patients clinical tool for treating NSCLC, as well as large and
in the Florida study receive 80 CGE at 2 CGE per irregular tumor volumes in other anatomic sites.
fraction to PET-positive deposits of gross primary
disease and PET-positive deposits of gross nodal
disease measuring more than 15 mm; 60 CGE at 2 Acknowledgments I would like to thank William Preston
EdD for manuscript editing and preparation.
CGE per fraction to PET-positive deposits of gross
nodal disease measuring less than 15 mm; and 40
CGE in 2 CGE per fraction to full nodal stations
containing foci of PET-positive gross disease, or References
anatomically adjacent to nodal stations containing
PET-positive gross disease. Bonnet RB, Bush D, Cheek GA, Slater JD, Panossian D, Franke
C, Slater JM (2001) Effects of proton and combined proton/
photon beam radiation on pulmonary function in patients
with resectable but medically inoperable non-small-cell
lung cancer. Chest 120:1803–1810
6 Conclusion Bush DA, Slater JD, Shin BB, Cheek G, Miller DW, Slater JM
(2004) Hypofractionated proton beam radiotherapy for stage
I lung cancer. Chest 126:1198–1203
Evidence shows that dose escalation can provide Byhardt RW, Martin L, Pajak TF, Shin KH, Emami B, Cox JD
improved local tumor-control in NSCLC, and treat- (1993) The influence of field size and other treatment factors
ment-planning comparisons reveal that proton treat- on pulmonary toxicity following hyperfractionated irradia-
ment can further reduce the dose to lung tissues even tion for inoperable non-small cell lung cancer (NSCLC).
Analysis of a radiation therapy oncology group (RTOG)
when compared to advanced photon treatments such
protocol. Int J Radiat Oncol Biol Phys 27:537–544
as IMRT and SBRT. Clinical results indicate that lung Chang JY, Zhang X, Wang X, Kang Y, Riley B, Bilton S,
cancer treatment with protons is feasible and has Mohan R, Komaki R, Cox JD (2006) Significant reduction
demonstrated a high level of local tumor-control for of normal tissue dose by proton radiotherapy compared with
three-dimensional conformal or intensity-modulated radia-
small lesions in reported trials. Severe treatment-
tion therapy in stage I or stage III non-small-cell lung
related toxicity has been minimal in both early and cancer. Int J Radiat Oncol Biol Phys 65:1087–1096
locally advanced cases, thus allowing for further dose Engelsman M, Kooy H (2005) Target volume dose considera-
escalation if clinically indicated. tions in proton beam treatment planning for lung tumors.
Med Phys 32:3549–3557
The evidence required to bring new technology
Engelsman M, Rietzel E, Kooy H (2006) Four-dimensional
into clinical practice is poorly defined. Historically, proton treatment planning for lung tumors. Int J Radiat
new technology that demonstrates improved dose Oncol Biol Phys 64:1589–1595
Proton Therapy for Lung Cancer: State of the Science 751

Fowler JF (2003) What can we expect from dose escalation cases of centrally and superiorly located stage I non-small-
using proton beams? Clin Oncol 15:S10–S15 cell lung cancer. Int J Radiat Oncol Biol Phys, epub ahead
Grills IS, Yan D, Martinez AA, Vicini FA, Wong JW, of print, doi: 10.1016/j.ijrobp.2010.03.012
Kestin LL (2003) Potential for reduced toxicity and dose Rosenman JG, Halle JS, Socinski MA, Deschesne K, Moore
escalation in the treatment of inoperable non-small-cell lung DT, Johnson H, Fraser R, Morris DE (2002) High-dose
cancer: a comparison of intensity-modulated radiation conformal radiotherapy for treatment of stage IIIA/IIIB
therapy (IMRT), 3D conformal radiation, and elective nodal NSCLC: technical issues and results of a phase I/II trial. Int
irradiation. Int J Radiat Oncol Biol Phys 57:875–890 J Radiat Oncol Biol Phys 54:348–356
Jemal A, Siegel R, Xu J, Ward E (2010) Cancer statistics, 2010. Rubin P, Casarett GW (1968) Clinical radiation pathology.
CA Cancer J Clin 60:277–300 W. B. Saunders, Philadelphia
Kong FM, Ten Haken RK, Schipper MJ, Sullivan MA, Chen M, Rubin P, Cooper RA, Phillips TA (eds) (1978) Radiation
Lopez C, Kalemkerian GP, Hayman JA (2005) High dose biology and radiation pathology syllabus. Am Coll Radiol
radiation improved local tumor control and overall survival Pub, Chicago
in patients with inoperable/unresectable non-small-cell lung Rubin P, Johnston CJ, Williams JP, McDonald S, Finkelstein JN
cancer: long-term results of a radiation dose escalation (1995) A perpetual cascade of cytokines postirradiation leads
study. Int J Radiat Oncol Biol Phys 63:324–333 to pulmonary fibrosis. Int J Radiat Oncol Biol Phys 33:99–109
Mountain CF (1997) Revisions in the international system for Saunders M, Dische S, Barrett A, Harvey A, Gibson D,
staging lung cancer. Chest 111:1710–1717 Parmar M (1997) Continuous hyperfractionated accelerated
Moyers MF, Miller DW, Bush DA, Slater JD (2001) radiotherapy (CHART) versus conventional radiotherapy in
Methodologies and tools for proton beam design for lung non-small cell lung cancer: a randomized multicentre trial.
tumors. Int J Radiat Oncol Biol Phys 46:1429–1438 Lancet 350:161–165
Murshed H, Liu HH, Liao Z, Barker JL, Wang X, Tucker SL, Shioyama Y, Tokuuye K, Okumura T, Kagei K, Sugahara S,
Chandra A, Guerrero T, Stevens C, Chang JY, Jeter M, Cox Ohara K, Akine Y, Ishikawa S, Satoh H, Sekizawa K (2003)
JD, Komaki R, Mohan R (2004) Dose and volume reduction Clinical evaluation of proton radiotherapy for non-small-
for normal lung using intensity-modulated radiotherapy for cell lung cancer. Int J Radiat Oncol Biol Phys 56:7–13
advanced-stage non-small-cell lung cancer. Int J Radiat Zhang X, Li Y, Pan X, Xiaoqiang L, Mohan R, Komaki R,
Oncol Biol Phys 58:1258–1267 Cox JD, Chang JY (2010) Intensity-modulated proton
Nakayama H, Sugahara S, Tokita M, Satoh H, Tsuboi K, therapy reduces the dose to normal tissue compared with
Ishikawa S, Tokuuye K (2010) Proton beam therapy for intensity-modulated radiation therapy or passive scattering
patients with medically inoperable stage I non-small-cell proton therapy and enables individualized radical radiother-
lung cancer at the University of Tsukuba. Int J Radiat Oncol apy for extensive stage IIIB non-small-cell lung cancer: a
Biol Phys 78:467–471 virtual clinical study. Int J Radiat Oncol Biol Phys 77:357–
Nihei K, Ogino T, Ishikura S, Nishimura H (2006) High dose 366
proton beam therapy for stage I non-small-cell lung cancer. Zhao L, Sandison GA, Farr JB, Hsi WC, Wu H, Li XA (2007)
Int J. Radiat Oncol Biol Phys 65:107–111 Patient-specific margins for proton therapy of lung. Australas
Register SP, Zhang X, Mohan R, Chang JY (2010) Proton Phys Eng Sci Med 30:344–348
stereotactic body radiation therapy for clinically challenging
Carbon Ion Radiotherapy
in Hypo-Fractionation Regimen and Single
Dose for Stage I Non-small-Cell Lung Cancer
T. Miyamoto, N. Yamamoto, M. Baba, and T. Kamada

Contents Abstract
It has been more than one decade since we started
1 Introduction.............................................................. 754 carbon ion radiation therapy (CIRT) for non-small-
2 Hypo-Fractionation Irradiation ............................. 755 cell lung cancer (NSCLC) in November 1994.
2.1 Materials and Methods .............................................. 755 From 1994 to 1999, we conducted a phase I/II
2.2 Results........................................................................ 756 clinical trial for stage I NSCLC with CIRT and
2.3 Discussion .................................................................. 757
demonstrated an optimal dose of 90 GyE in 18
2.4 Conclusions................................................................ 758
fractions over 6 weeks and 72 GyE in 9 fractions
3 Single-Dose Irradiation ........................................... 759 over 3 weeks for achieving more than 90% local
3.1 Patients and Treatment.............................................. 759
3.2 Staging ....................................................................... 759 control with minimal pulmonary damage. In the
3.3 Results and Discussion.............................................. 759 following phase II study from 1999 to 2003, the
total dose was fixed at 72 GyE in 9 fractions over
References.......................................................................... 761
3 weeks and at 52.8 GyE for stage IA and at
60 GyE for stage 1B in 4 fractions over 1 week.
Targets were irradiated from four oblique direc-
tions. A respiratory-gated irradiation system was
used for all irradiation sessions. On these two
phase II schedules combined, the 5-year local
control rate for 131 primary tumors of 129 patients
was 91.5%. The local control rate for T1 and T2
tumors was 96.3 and 84.7%, respectively. While
there was significant difference in control rate
between T1 and T2, there was no significant
difference in histology between squamous and
non-squamous type. The 5-year cause-specific
survival rate of the patients was 67.0% (IA: 84.4,
IB: 43.7), and their overall survival was 45.3%
(IA: 53.9, 1B: 34.2). No adverse effects greater
than grade III occurred in the lung. In this way, the
treatment period and fractionation were shortened
and lessened from 18 fractions over 6 weeks to 9
T. Miyamoto (&)  N. Yamamoto  M. Baba  T. Kamada fractions over 3 weeks and further to 4 fractions
Research Center Hospital for Charged Particle Therapy,
National Institute of Radiological Sciences (NIRS), over one week. Finally it reached a single dose.
4-9-1 Anagawa, Inage-Ku, Chiba 263-8555, Japan Since 2003, 210 patients have already been treated
e-mail: t-miyamoto@min-iren-c.or.jp

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 753
DOI: 10.1007/174_2011_257, Ó Springer-Verlag Berlin Heidelberg 2011
754 T. Miyamoto et al.

with CIRT in single dose increasing 28, 32, 34, 36, Table 1 Results of phase I/II study on CIRT for stage I
38, 40, 42, 44, 46, and 48 GyE. Compared with the non-small-cell lung cancer
previous fractionation regimen, CIRT in single- Adverse effects on lung
dose is demonstrating low morbidity and high (1) Minimum damage on lung (Grade 3 radiation pneumonitis
QOL. The 5-year local control rate of 131 tumors was 2.7%)
with doses more than 36 GyE was higher than (2) Influenced by dose, respiration movement, and port
80%. The 5-year cause-specific and overall sur- direction and number
vival rate of 131 patients were 1.5 and 52.6%, Local control
respectively. Of the whole evaluate, we will finally (1) Dose dependent, but less dependent to tumor size and
recommend that CIRT in single dose is the best for histological type
the treatment of the peripheral type of stage I (2) More than 90% by optimal dose and evidenced by
NSCLC. pathological CR
Survival
(1) Influenced by local control state and tumor size
(2) Less decreased by node and intralober (PM1) metastasis
1 Introduction (3) More decrease by local failure, pleuritis carcinomatosa
and distant metastasis
In 1998, lung cancer became the leading cause of
cancer-related death in Japan, as in the Western
countries. Surgery plays a pivotal role in the curative For clinical use, carbon ion beams were selected
treatment for non-small-cell lung cancer (NSCLC), based on preclinical experiments (Kanai et al. 1999).
but it is not necessarily the best treatment for elderly Dose escalation is essential to improve the effec-
persons and/or patients with cardiovascular and pul- tiveness of carbon ion radiation therapy (CIRT) for
monary complications. Conventional radiotherapy as lung cancer. Yet this increases the risk of pulmonary
an alternative, however, produces a 5-year survival toxicity. The phase I/II clinical trials with CIRT for
rate in merely 10–30% (Jeremic et al. 2002) of the stage I NSCLC were initiated in October 1994 and
patients treated due to poor control of the primary concluded in February 1999. The additional purpose
tumor. These results were quite inferior to those by was to develop correct, reliable, and safe irradiation
surgery (Mountain 1997; Naruke et al. 2001) techniques for CIRT. As a result of two successive
Recently, new modalities such as stereotactic radio- dose escalation phase I/II studies, a regimen of
therapy (Nagata et al. 2005; Onishi et al. 2004) and 90 GyE (Gray equivalents) in 18 fractions over
proton radiotherapy (Bush et al. 1999; Shioyama et al. 6 weeks (NIRS protocol number: #9303) and 72 GyE
2003) have been tried but have not yet been demon- in 9 fractions over 3 weeks (#9701) were chosen as
strated as valid alternatives. the optimal dose (Miyamoto et al. 2003), and the
Heavy particle beams penetrate the body and following results (Table 1) were obtained: (1) The
abruptly stop to form a Bragg peak as energy is lost; local control rate was dose dependent, and reached
A great deal of energy is deposited to cause dense more than 90% at 90 GyE with a regimen of 18
ionization of high linear energy transfer (LET) (Kanai fractions over 6 weeks and 72 GyE with a regimen of
et al. 1997). These physical properties help achieve 9 fractions over 3 weeks. Both the doses were deter-
excellent dose-localization for deep-seated tumors, mined to be optimal. It was found that setting the
sparing critical normal organs while high LET exerts provisional target by allowing for the difference with
a powerful biological effect. It can be expected that CT value can prevent marginal recurrence (Koto et al.
heavy particle beams can improve tumor control 2004). (2) The damage on lung was minimum,
without increasing toxicity. showing that Grade 3 radiation pneumonitis occurred
In 1984, the Japanese government decided to in 2.7%. Respiratory-gated and 4-portal oblique irra-
construct the Heavy Ion Medical Accelerator in Chiba diation excluding opposed ports proved successful in
(HIMAC) at our Institute: National Institute of reducing the incidence of radiation pneumonitis.
Radiological Sciences (NIRS). HIMAC was com- (3) The survival was strongly influenced by the
pleted in November 1993 (Hirao et al. 1992). local control and tumor size in the primary lesion.
Carbon Ion Radiotherapy in Hypo-Fractionation Regimen and Single Dose 755

(4) The early detection of nodal and intralobar The carbon ion radiation dose was expressed in
metastasis followed by irradiation with carbon beams terms of Gray equivalents (GyE). The GyE values are
can prevent the survival rate from further decreasing. calculated by multiplying the physical dose with the
(5) The local failure, distant metastasis, and pleuritis relative biological effectiveness (RBE), which is
carcinomatosa caused to decrease the survival. approximately 3.0 at 0.8 cm from the distal end of the
SOBP (Kanai et al. 1997, 1999).

2 Hypo-Fractionation Irradiation 2.1.2 Patients


Patients with a peripheral type of stage I NSCLC who
Following the phase I/II study, we have conducted were inoperable or refused surgery were eligible for
two successive phase II studies: the total dose was this study. Primary tumor had to be histologically
fixed at 72 GyE in 9 fractions over 3 weeks (#9802) proven and measurable. The patient’s performance
(Miyamoto et al. 2007a, b) and at 52.8 GyE for stage status (PS) was between 0 and 2 according to the
IA and at 60 GyE for stage IB 4 fractions over one WHO criteria. The patient had to have no history of
week (#0001) (Miyamoto et al. 2007a, b). Using the radiotherapy to the target, and no prior chemotherapy
two hypo-fractionation regimen, the phase II trial was within 4 weeks of therapy. They had to have no
initiated in April in 1999 and closed in December in intractable infection and active multiple cancers. Each
2003 accruing a total number of 129 patients. patient gave his/her written informed consent,
approved by the Ethics Committee at our Institute.
After therapy, the patients’ progress was verified
2.1 Materials and Methods twice a year by the members of the Working Group
for Lung Cancer.
2.1.1 Carbon Ion Beams, HIMAC One hundred and twenty nine patients with 131
and Treatment Systems primary lesions were enrolled into this study. The 51
Carbon ion beams with 290, 350 and 400 MeV/ primary tumors of 50 patients were treated by carbon
nucleon energy were generated in the HIMAC syn- beam irradiation alone using the fixed total dose of
chrotron and delivered through its transport system to 72 GyE in 9 fractions over 3 weeks (#9802). The
the irradiation system in the treatment room. In this remaining 79 patients had 80 stage I tumors. The IA
system, the beams are broadened and shaped to a stage tumors were treated with the fixed dose of
three-dimensional (3-D) tumor contour. For 3-D dose 52.8 GyE and the IB tumors with the fixed does of
delivery, the key technology is to spread out a sharp, 60 GyE in 4 fractions over one week (#0001). The
monoenergetic Bragg peak (SOBP) of carbon ion patients had an average age of 74.5 years. The gender
beams by using a range modulator to cover the tumor breakdown was 92 males and 37 females. The tumors
thickness (Kanai et al. 1997, 1999). The reference consisted of 72 of T1 and 59 of T2. The average
point was set so as to be the center of SOBP. In this tumor size was 31.5 mm in diameter. By type of
procedure, the patients were immobilized with cus- cancer, there were 85 adenocarcinomas, 43 squa-
tom-made devices in a semi-cylindrically shaped mous cell carcinomas, 2 large cell carcinomas and
rotary capsule set on a CT couch or a treatment couch. 1 adenosquamous cell carcinoma. Medical inoper-
CT planning is performed using the HIPLAN (Endo ability stood at 76% (Table 2).
et al. 1996), which was specifically developed for 3-D
treatment planning with respiratory-gated CT images. 2.1.3 Treatment
Patient positioning and verification are performed The targets were usually irradiated from four oblique
using the patient set-up devices, digital reconstructed directions without prophylactic elective nodal irradi-
radiographs (DRR), online portal fluoroscopic radio- ation (ENI). A greater than 10 mm margin was set
graphs and metal markers made of iridium-wire as a outside gross target volume (GTV) to determine the
landmark. A respiratory-gated irradiation system was clinical target volume (CTV). The planning target
developed and employed to minimize respiratory volume (PTV) was set by adding an internal margin
movements of the tumor and reduce treatment volume (IM) to the CTV. The IM was determined by extend-
(Minohara et al. 2000). ing the target margin in the head and tail directions by
756 T. Miyamoto et al.

Table 2 Treatments and Characteristics of 129 patients with stage 1 NSCL (1999.4–2004.9)
Protocol no 9802 0001 Total
Fractionation total dose (GyE) 9fr/3w 72 4fr/1w 52.8 (T1), 60 (T2) –
Patient number 50 79 (77)* 129 (127)*
Lesion number 51 80 131
Age 74 75 74.5
Gender
Male 38 54 92
Female 12 25 37
PS
0 0 8 8
1 50 68 118
2 0 3 3
T factor (stage)
T1 (IA) 30 42 72
T2 (IB) 21 38 59
Tumor size (mm) 30.9 32.1 31.5
Histology
AD 32 53 85
SQ 19 24 43
L 0 2 2
Ad-Sq 0 1 1
Refusal of surgery 14 17 31 (24%)
Medical inoperability 36 62 98 (76%)
*The two patients were twice treated in the protocol firstly 9802 and the 0001 secondly

a width of 5 mm, resulting in the successful preven- (non-Sq) (n = 88) was 87.1 and 93.8%, respectively.
tion of marginal recurrence caused by respiration While there was significant difference (p = 0.0156)
movement (Koto et al. 2004). Figure 1 shows the dose in tumor control rate between T1 and T2, there was no
distribution map for a representative case. significant difference (p = 0.1516) between squa-
mous and non-squamous in T1 ? T1, and in the both
2.1.4 Statistical Analysis T1 and T2. However, with respect to squamous cell
Local control and survival were assessed by the type the local control was 100% for T1 (n = 17) and
Kaplan–Meier method. For statistical testing, the 78.0% for T2 (n = 26), and there was near significant
long-rank test was used. difference (p = 0.0518). The local control of non-
Squamous tumors was 95.3% for T1 (n = 55), and
91.0% for T2 (n = 33), and there was no significant
2.2 Results difference (p = 0.3364).
The 5-year cause-specific survival rate of the 129
All patients were followed until death with a median patients was 67% (Fig. 2), breaking down into 84.8%
follow-up time of 50.8 months ranging from for the stage IA and 43.7% for the stage IB tumors.
2.5 months to 70.0 months. The local control rate for The 5-year overall survival rate was 45.3% (Fig. 2),
the 131 primary lesions was 91.5% (Fig. 2). The local breaking down into 53.9% for the stage IA and 34.2%
control rate for the T1 (n = 72) and T2 (n = 59) for the stage IB tumors.
tumors was 96.3 and 84.7% and for squamous The toxicity of CIRT to the skin and lung
cell type (Sq) (n = 43) and non-squamous cell type was assessed according to the RTOG (acute) and
Carbon Ion Radiotherapy in Hypo-Fractionation Regimen and Single Dose 757

Fig. 1 a Axial image: dose


distribution for tumor (right
lower lobe) receiving a total
carbon ion dose of 60 GyE
from 4 directions (4-portals).
Yellow line: PTV red line:
96% of the prescribed dose
orange line: 90%, green line
50%, blue line 30%, and
violet line: 10%. b Coronal l
image. c Sagittal image

Local Control (n=131)& Survival(n=129) 126 patients were at grade 1 and 3 patients were at
1.0 grade 2. No more than grade 3 reactions were
Local Control and Survival rate

Local Control (5y) 91.5%

Cause-specific(5y) 67.7%
observed.
0.8
The cause of death was assessed. Sixty-two out of
0.6 Overall (5y) 45.3% 129 patients (48.8%) were dead at that time, with 31
dying of the lung tumor treated with CIRT. Five of
0.4
the patients died due to primary recurrence and 26
0.2 patients due to metastasis and dissemination. For the
remaining 31 patients, intercurrent diseases were the
0.0
cause of death.
0 10 20 30 40 50 60
Month after treatment

Fig. 2 Five-year local control (blue), cause-specific (red), and 2.3 Discussion
overall (green) survivals in 129 patients with 131 stage I
NSCLC after start of CIRT on the following two phase-II In the present study, we used the fixed total dose of
regimen combined: the total dose was fixed at 72 GyE in 9 72 GyE in 9 fractions over 3 weeks, and of 52.8 GyE
fractions over 3 weeks (#9802) and at 52.8 GyE for stage IA
and at 60 GyE for stage 1B in 4 fractions over 1 week (#0001) for stage IA and 60 GyE for stage IB 4 fractions over
one week. Using this schedule which was optimized
RTOG/EOTRC (late) as shown in Table 3. The early in the phase I/II study by CIRT (Miyamoto et al.
skin reaction was assessed for 131 lesions and the late 2003), we conducted a phase II clinical trial for 129
skin reactions for 128 leisions. In acute reaction 125 patients with stage I NSCLC.
lesions were at grade 1 and 6 at grade 2. In late As a result, the local control, cause-specific, and
reaction 126 lesions were at grade 1, one at grade 2 overall survival rates for 129 patients were 91.5, 67.0,
and one at grade 3. The lung reaction was clinically and 45.3%, respectively. And toxicity in skin, lung
assessed for 129 patients. There were 127 patients at and bone was of a minimum.
grade 0 and 2 patients at grade 2 in early reaction. Local recurrence out of 131 primary cancers of
There were 126 patients who were followed up for 129 patients occurred in 9 cases (6.8%). The average
late effect. Among them, 7 patients were at grade 0, recurrence time was 17.2 months in an average
758 T. Miyamoto et al.

Table 3 Adverse effects by CIRT


Early reaction (RTOG) Late reaction (RTOG/EORTC)
Lesion no 0 1 2 3 4B Lesion no 0 1 2 3 4B
Skin 9802 51 0 50 1 0 0 51 0 49 1 1 0
0001 80 0 75 5 0 0 77* 0 77 0 0 0
Total 131 0 125 6 0 0 128 0 126 1 1 0
Lung 9802 50 49 0 1 0 0 50 0 48 2 0 0
0001 79 78 0 1 0 0 76* 7 68 1 0 0
Total 129 127 0 2 0 0 126 7 116 3 0 0
*3 cases were not observed due to the early death

ranging from 7 to 39 months. According to the pre- dissection had adverse effects on recurrence and sur-
vious study, the observation period required to vival. On the contrary, our treatment strategy for
determine the local control of the irradiated lesions regional recurrence is thought to have been validated
was at least 3 years after therapy. However, the as the standard surgical procedure for stage I NSCLC.
present study suggests that a longer observation time With clinical stage I NSCLC, our 5-year overall
is needed. Prolonged survival guarantees more reli- survival results were somewhat inferior to the surgical
able observation of local control results. ones (Miyamoto et al. 2007a, b; Yamamoto et al.
For the correct assessment of local control of the 2003). The survival difference may be due to age
patients who could not be observed for such a long difference between the two groups. The incidence of
time as they died of metastasis/dissemination or death due to recurrence in the surgical group was 29
intercurrent disease, a histological approach by or 36% whereas that of death due to intercurrent
repeating bronchoscopy was performed and proved to diseases was 19% or a few % (Martini et al. 1995). On
be no viable tumor cells in the collected specimens the other hand, there was more frequent intercurrent
(Miyamoto et al. 2007a, b). And this definite tumor death (60%) than recurrence death (40%) with our
control by CIRT was also observed in autopsy and patients. Generally speaking, such a high frequency of
operated cases (Yamamoto et al. 2003). Such high intercurrrent death might be due to the advanced age
and definite tumor control appears to be an out- of our patients who were 10 years older on average
standing feature of CIRT which may be primarily due than the surgical patients.
to the radiobiological nature of the high LET beams, Compared with pulmonary damage reported in
and may contribute to a higher survival rate for stage I stereotactic radiotherapy for stage I NSCLC (Nagata
NSCLC. The failure in local control for a primary et al. 2005; Onishi et al. 2004), the incidence and
tumor directly influences the poor survival of stage I severity in our patients seem to be clearly low. These
NSCLC patients (Miyamoto et al. 2003, 2007a, b). less adverse effects for the lung were achieved as a
There were 20 loco-regional recurrences (15.7%): result of the small irradiated volume (V20 #9802) T1
7 regional nodes, 1 intrabronchial metastasis and 3 (n = 30) mean 5.5% (2.3–11.6), T2 (n = 39) mean
intralobar pulmonary metastases (PM1) in addition to 6.4% (1.0–12.3), #0001 T1 (n = 41) mean 4.8%
9 primary recurrences. Their incidence was close to (1.1–13.2) T2 (n = 21) mean 7.6% (2.6–13.9)
that of surgery (7.5%) (Harpole et al. 1995), 11% achieved with excellent dose distribution by carbon
(Martini et al. 1995). When the 11 regional metastasis ion beams due to the formation of a Bragg peak
were diagnosed after the first CIRT for a primary contrast with X-ray as a permeating beam.
tumor, the second CIRT was given to the 9 regional
recurrences and photon therapy to two recurrences.
All of the regional recurrences could be controlled by 2.4 Conclusions
re-treatment irradiation, and the patients were found
to have the same survival as the patients without By conducting the phase II study using a total dose of
primary recurrence. Martini et al. (1995) reported any 72 GyE in a regimen of 9 fractions over 3 weeks
resections less than lobectomy and no lymph node and 52.8 GyE for stage IA and 60 GyE for stage IB
Carbon Ion Radiotherapy in Hypo-Fractionation Regimen and Single Dose 759

4 fractions over one week the following results were patients shown in Table 2. Irradiation is performed in
obtained: (1) The higher local control rate of 91.5% B room equipped with vertical and horizontal ports. A
was achieved. In addition, there was statistical diff- patient immobilized in rotary capsule is rotated at +20
erence in the local control rate for T1 and T2 and near and -20° and irradiated in order from each port
significant for squamous cell carcinoma and non- within 15 min, and completed from 4-ports within 1 h
squamous cell carcinoma of T2. (2) The damage on and a case was shown after therapy in Fig. 3. As
lung was minimum, showing no Grade 3 radiation phase I/II study, a dose of 28 GyE for 6 patients (pts),
pneumonitis. (3) The cause-specific survival rates 32 GyE for 27 pts, 34 GyE for 34 pts, 36 GyE for 18
were equal to that of surgery while the overall sur- pts, 38 GyE for 14 pts, 40 GyE for pts 20, 42 GyE for
vival was less than that of surgery due to high inci- 15 pts, 44 GyE for 44 pts, and 46 GyE for 20 pts was
dence in intercurrent death due to an advanced age delivered.
and complication. CIRT, which is an excellent new
modality in terms of a high QOL and ADL, is a valid
alternative to surgery for stage I cancer, especially for 3.2 Staging
elderly and inoperable patients.
For staging, CT scan of the chest and upper abdomen,
enhanced MRI scans of the brain, bone scans and
3 Single-Dose Irradiation bronchoscopy were routinely performed. Regional
lymph nodes greater than 1 cm in the short axis on the
Throughout the 10 years of CIRT for clinical stage I contrast-enhanced CT images were taken to serve as
NSCLC as already described, the treatment period evidence of positive metastasis. A 11C-Methionine
and fractionation were shortened and lessened from PET scan was routinely taken for confirmation as
18 fractions over 6 weeks to 9 fractions over 3 weeks previously (Yasukawa et al. 1996). However, there
and further to 4 fractions over one week. Finally it were some false positive cases by PET scan. Since
reached a single dose (Miyamoto 2004). Till now, 210 2005, we adopted EBUS (endobronchial ultrasound-
patients have already been treated with single-dose guided transbronchial needle aspiration) to exclude
CIRT (#2010) with doses increasing 28, 32, 34, 36, the false positive node by PET scan (Nakajima et al.
38, 40, 42, 44, 46, 48 and 50 GyE since April 2003. 2008).
The single dose CIRT therapy has been demonstrated
to lead to low morbidity and a high QOL. The efficacy
of single dose CIRT close to that of the previous 3.3 Results and Discussion
fractionation regimen with increasing dose and
attained the same level of local control and survival For adverse effects, there was no grade III for early
rate with doses more than 36 GyE. (198 pts) and late (128 pts) lung reaction and in early
(198 pts) and late (198 pts) skin reaction. The 5-year
local control rate, cause-specific and overall survival
3.1 Patients and Treatment of 131 patients who received a dose of 36–46 GyE
was 80.5, 71.5, and 52.6%, respectively as shown in
Patient eligibility was same as that of the previous Fig. 4. The local control rate of T1 (78 patients) and
fractionation regimen. Until the present time, 201 T2 (53 patients) was 86.7 and 78.4% and three years
patients were treated with single-dose CIRT. Among later, it was 82.8 and 78.4%, respectively. This study
them, 198 patients who had elapsed 6 months after is on-going, now. These results are further improving
therapy were documented for analysis. An average with increasing dose. Recently, we could determine
age was 74.2 year-old. The number of male to female an optimal single-dose of CIRT for the treatment of
was 144–54. There were 134 adenocarcinoma, 62 peripheral type of stage I NSCLC, and all of the
squamous cell carcinoma, 3 large cell carcinoma and results will be published.
one mucoepitheloid cancer. The number T1 and T2 The massive single dose technique for uterine
was 108 and 85, respectively. These characteristics of cancer was first initiated in the 1920s in Germany
patients were almost same to those of the previous after the First World War (Ludwig Seitz and Hermann
760 T. Miyamoto et al.

Fig. 3 In B room equipped with vertical and horizontal ports, upper left and middle). Dose-distribution curve for stage IB
a patient immobilized in rotary capsule is rotated at +20° and adenocarcinoma of the lung treated with single-dose CIRT
-20° and irradiated in order from each port within 15 min. The from four oblique directions (the right) and the tumor response
irradiation was completed from 4-ports within one hour (the (the lower) before and after 6 years at irradiation was shown

Wintz 1920). In an attempt to concentrate on the by using a drawn-out dose fractionation regimen with
tumor control dose in the cancer, Ludwig Seitz and few adverse effects. The present standard technique is
Hermann Wintz used a method involving irradiation a fractionation regimen in which the required radia-
from multiple portals to improve spatial dose distri- tion dose is spread into fractions over a certain period
bution and control the radiation dose to within the of time. The goal is to achieve the highest possible
tolerance dose of the normal tissue. This method is local control within the permissible range of radiation
called the Erlangen technique that constitutes the injury, especially late injuries. However, this is a
basic rationale for present-day radiotherapy. Since the substantially palliative method of radiotherapy. Now
beam used at the time had a poor depth/dose ratio and that we have been able to apply carbon ion beams that
the dose measurement techniques available were still have a superior depth dose distribution for radio-
not sufficiently sophisticated, visceral injuries and therapy and select the treatment for stage I lung
other side-effects occurred quite frequently. After cancer, our single-dose regimen is a revival of the
this, improvements have been made until the present technique using a heavy particle beam created as the
Carbon Ion Radiotherapy in Hypo-Fractionation Regimen and Single Dose 761

at more than 36.0GyE (n=131) Acknowledgments We thank the following doctors who par-
ticipated in this long-term study: Hideki Nisimura, Masasi
1 Koto, Toshiyuki Sugawara, Tomoyasu Yasiro, Naoki Hirasawa,
Local Control and survival rate

Kenji Kagei, Mio Nakajima, Toshio Sugane, Kyousan Yosik-


.8 awa, Susumu Kandatsu, Hidefumi Ezawa and Kennosuke
Kadono. We also thank the doctors who constantly supported
.6 and advised: Hirohiko Tujii, Jun-etsu Mizoe, Suhou Sakata,
Kozo Morita, Takeshi Iinuma, and Toru Matzumoto, and
.4 Takehiko Fujisawa of the Working group for lung cancer.

.2 Local control(5 years) : 80.5%


Cause-specific (5 years) : 71.5% References
0 Over all (5 years) : 52.6%

0 1 2 3 4 5 6 ACRIN (2010) For immediate release. Screening of people at


Years after treatment high-risk for lung cancer with low dose CT significantly
reduces lung cancer death
Fig. 4 Five-year local control (blue), cause-specific (red), and Bush DA, Slater JD, Bonnet R et al (1999) Proton-beam
overall survivals (green) in 131 patients with 131 stage I radiotherapy for early stage lung cancer. Chest
NSCLC after start of single-dose CIRT (#2010) with doses 116:1313–1319
more than 36 GyE Endo M, Koyama-Ito H, Minohara S et al (1996) Hiplan-a
heavy ion treatment planning system at HIMAC. J Jpn Soc
Ther Radiol Oncol 8:231–238
Harpole DH, Herndon JE, Yung WG et al (1995) Stage I nonsmall
result of modern science and we are confident that it cell lung cancer. A multivariate analysis of treatment methods
will mark the beginning of a new era of radiotherapy and patterns of recurrence. Cancer 76:787–796
for the 21st century. To achieve this goal we will Hirao Y, Ogawa H, Yamada S et al (1992) Heavy ion
synchrotron for medical use—HIMAC project at NIRS-
remain committed to further research for various JAPAN. Nucl Phys A 538:541c–550c
cancers at early stage. Jeremic B, Classen J, Bamberg M (2002) Radiotherapy alone
Irradiation in our single-dose CIRT is completed technically operable, medically inoperable, early-stage (I/II)
within one hour. By reducing the burden on the non-small-cell lung cancer. Int J Radiat Oncol Biol Phys
54:119–130
patient’s mental and physical, it is suitable for out- Kanai T, Furusawa Y, Fukutsu K et al (1997) Irradiation of
patient radiation and accordingly economical. mixed beam and design of spread out Bragg peak for heavy-
Reducing mortality due to lung cancer deaths is the ion radiotherapy. Radiat Res 147:78–85
most important issue. As a practical solution we Kanai T, Endo M, Minohara S et al (1999) Biophysical
characteristics of HIMAC clinical irradiation system for
propose a new paradigm for the fight against the lung heavy-ion radiation therapy. Int J Radiat Oncol Biol Phys
cancer (Iinuma and Miyamoto 2006). This approach 44:201–210
calls for the popularization of lung cancer screening Koto M, Miyamoto T, Yamamoto N, et al (2004) Local control
CT to cover over 50% of the entire Japanese popu- and recurrence of stage I non-small cell lung cancer after
carbon ion radiotherapy. Radiother Oncol 71:147–156
lation aged between 40 and 84 by 2025 and the Ludwig Seitz, Hermann Wintz (1920) Unsere Methode der
treatment of the early-lung cancer thus detected by Rontgen-Tiefentherapie und ihre ihre Erforge. Verlag von
screening CT with single-dose CIRT. By this means it Uaban & Schwarzenberg, Berlin N24, Wien I, 1920
would be possible to reduce the lung cancer mortality Martini N, Bains MS, Burt ME et al (1995) Incidence of local
recurrence and second primary tumors in resected stage I
by over 22%. The strategic installation of CIRT lung cancer. J Thorac Cardiovasc Surg 109:120–129
facilities exclusively dedicated to the treatment of Minohara S, Kanai T, Endo M et al (2000) Respiratory gated
lung cancer throughout Japan would permit reduction irradiation system for heavy ion radiotherapy. Int J Radiat
of lung cancer death on a large scale at a lower cost Oncol Biol Phys 47:1097–1103
Miyamoto T (2004) Carbon beam therapy for lung cancer. Jpn J
than with current surgery. Fortunately, NCI Lung Cancer 44:741–751
announced in November 2010 (ACRIN 2010) that Miyamoto T, Yamamoto N, Nishimura H et al (2003) Carbon
a large-scale RCT for lung cancer screening by ion radiotherapy for stage I non-small cell lung cancer.
low-dose CT demonstrated the reduction of lung Radiother Oncol 66:127–140
Miyamoto T, Baba M, Yamamoto N, Koto M et al (2007a)
cancer death by 20% and total death by 7% of for Curative treatment of stage I non-small cell lung cancer
heavy smokers. with carbon ion beams using a hypo-fractionated. Int J
Our proposal will come true in the near future. Radiat. Oncol Biol Phys 67:750–758
762 T. Miyamoto et al.

Miyamoto T, Baba M, Yamamoto N, Sugane T et al (2007b) Onishi H, Araki T, Shirato H et al (2004) Stereotactic
Carbon ion radiotherapy for stage I non-small cell lung hypofractionated high-dose irradiation for stageI nonsmall
cancer using a regimen of four fraction during 1 week. cell lung carcinoma. Cancer 101:1623–1631
J Thorac Oncol 2:916–926 Shioyama Y, Tokuuye K, Okumuma T et al (2003) Clinical
Mountain CF (1997) Revisions in the international system for evaluation of proton radiotherapy for non-small-cell lung
staging lung cancer. Chest 111:1710–1717 cancer. Int J Radiat Oncol Biol Phys 56:7–13
Nagata Y, Takayama K, Matuo Y et al (2005) Clinical Iinuma T, Miyamoto T (2006) A new paradigm for medical
outcomes of a phase I/II study of 48 Gy of stereotactic body practice for lung cancer: A combination of lung cancer
radiotherapy in 4 fractions for primary lung cancer using a screening by LSCT (lung cancer screening CT) and radio-
stereotactic body frame. Int J Radiat Oncol Biol Phys surgery by single-dose carbon irradiation. JJLC 46:309–314
63:1427–1431 Yamamoto N, Miyamoto T, Nishimura H et al (2003)
Nakajima T, Yasufuku K, Fujiwara T et al (2008) Endobron- Preoperative carbon ion radiotherapy for non-small cell
chial ultrasound-guided transbronchial needle aspiration for lung cancer with chest wall invasion-pathological findings
the diagnosis of intrapulmonary lesions. J Thorac Oncol concerning tum response and radiation induced lung injury
3(9):985–988 in the resected organs. Lung Cancer 42:87–95
Naruke T, Tsuchiya R, Kondo H et al (2001) Prognosis and Yasukawa T, Yamaguchi Y, Aoyagi H et al (1996) Diagnosis of
survival after resection for bronchogenic carcinoma based hilar and mediastinal lymph node metastasis of lung cancer
on the 1997 TNM-staging classification: the Japanese by positoron emission tomography using 11C-methionine.
experience. Ann Thorac Surg 71:1759–1764 Jpn J Lung Cancer 36:919–926
Novel Cytotoxic Agents in Combination
with Radiation in the Management
of Locally Advanced Non-Small Cell
Lung Cancer: Focus on Pemetrexed
and Nab-Paclitaxel [Abraxane]
Corey J. Langer

Contents Abstract
The treatment of locally advanced, unresectable
1 Introduction.............................................................. 765 non-small cell lung cancer (NSCLC) remains
2 Pemetrexed ............................................................... 766 challenging. Radiation therapy (RT) combined with
chemotherapy is more effective than RT alone, and
3 Abraxane................................................................... 768
concomitant chemoradiation has yielded improved
4 Conclusions ............................................................... 770 survival compared to sequential chemotherapy and
References.......................................................................... 770 RT, but at the cost of heightened toxicity, especially
esophagitis. The majority of randomized chemora-
diation trials have featured cisplatin or carboplatin
based chemotherapy, usually in combination with
either etoposide or paclitaxel, with median survival
times of 16–18 months, and 5 year survival rates of
15 to 20% at best. In this chapter, we describe recent
studies employing pemetrexed and nab-paclitaxel,
each of which has been extensively investigated in
advanced disease, the former yielding improved
progression-free and overall survival in adenocarci-
noma both in the frontline and maintenance setting,
the latter yielding superior response rates in
squamous cell carcinoma compared to conventional
paclitaxel in chemo-naïve patients. Both agents have
demonstrated safety and efficacy in LA-NSCLC in
conjunction with RT. Whether these agents will ever
prove superior to either etoposide or paclitaxel in this
setting will require careful, phase III testing.

1 Introduction
C. J. Langer (&) In fit patients with minimal weight loss (i.e., less
Abramson Cancer Center,
than 5–10% from baseline) and intact PS (ECOG PS
University of Pennsylvania,
Philadelphia, PA 19104, USA 0-1), concurrent chemoradiation with a platinum-
e-mail: corey.langer@uphs.upenn.edu based combination has emerged as the standard

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 765
DOI: 10.1007/174_2011_314, Ó Springer-Verlag Berlin Heidelberg 2011
766 C. J. Langer

approach in locally advanced NSCLC (Furuse et al.


1999; Curran et al. 2010; Aupérin et al. 2007). In the 2 Pemetrexed
absence of significant comorbidity, hearing loss, or
renal compromise, in patients who can readily tol- Based on a phase III trial showing equivalent
erate an acute fluid load, the use of the etoposide– response rate, PFS, and survival with considerably
cisplatin combination with XRT, both started on less toxicity compared to docetaxel, pemetrexed, a
day 1, is generally favored. The minimum ‘‘accep- synthetic, multi-targeted antifol, was first approved in
ted’’ dose of radiation in this setting is 60 Gy, the second line setting in advanced NSCLC (Hanna
although multiple studies are now evaluating doses et al. 2004). More recently, it was approved for first-
of 74 Gy and higher using newer technology. In line treatment in advanced non-squamous NSCLC
frailer or older patients, and in those with significant based on a phase III trial showing a survival advan-
co-morbidity including renal insufficiency (creati- tage for pemetrexed–cisplatin compared to gemcita-
nines of 1.5–3.0), hearing loss, congestive heart bine–cisplatin (Scagliotti et al. 2008). In addition, the
failure, or severe COPD, cisplatin is often discarded JMEN study demonstrated a >5 months survival
in favor of carboplatin, and many will substitute advantage for maintenance therapy with pemetrexed
paclitaxel for etoposide. Choy et al. (2002) investi- in a similar population of non-squamous cell NSCLC
gated regimens featuring carboplatin AUC 2 weekly compared to intravenous placebo in patients whose
and paclitaxel 45–50 mg/m2 weekly, both initiated disease had stabilized or responded to first-line
on day 1 of thoracic radiation, followed by two treatment with a platinum-based combination
cycles of full-dose chemotherapy once radiation was (Ciuleanu et al. 2009). As a result of these studies, use
completed; and Movsas et al. (2005) assessed this of pemetrexed in advanced NSCLC has expanded
approach in the cooperative group setting. globally.
Many have argued that carboplatin-based therapy Pre-clinical data suggest additivity, if not synergy
is inferior to cisplatin in the treatment of LA-NSCLC, for pemetrexed with radiation. Due to its effective-
but recent data from Japan in a combined modality ness, ease of administration, and relatively mild tox-
trial [West Japan Oncology Group Trial (WJTOG icities observed in advanced non-small cell lung
0105)] evaluating various concurrent chemoradiation cancer, pemetrexed has also been explored in locally
regimens suggests de facto therapeutic equivalence advanced NSCLC. A study led by Seiwert et al.
(Yamamoto et al. 2010). Investigators led by (2007) was among the first to assess the role of
Nobuyuki Yamamoto compared their erstwhile stan- pemetrexed and platinum in combination with radia-
dard of MVP with weekly carboplatin in combination tion in locally advanced NSCLC. As a phase I effort,
with either irinotecan or paclitaxel during XRT; in this study included patients with oligo-metastatic
each arm, those without disease progression or unto- disease as well as esophageal cancer. Treatment was
ward toxicity went on to receive two cycles of full- administered every 21 days. Regimen 1 included
dose chemotherapy during the ‘‘consolidation’’ period pemetrexed (200–600 mg/m2), while regimen 2 fea-
using the same agents administered during XRT. The tured pemetrexed (500 mg/m2) with escalating car-
paclitaxel–carboplatin regimen resulted in less toxic- boplatin doses (AUC = 4 ? 6). Both regimens
ity, fewer dose reductions or omissions, and equiva- included concurrent radiation, escalated from 40 to
lent, if not superior, survival at 5 years: 19.5% versus 66 Gy. Thirty patients (18 with locally advanced
17.5% for MVP and 17.8% for irinotecan/carboplatin. NSCLC and 12 metastatic with dominant local
In fairness, this study also compared second-gen- symptoms) were enrolled. All but one had an ECOG
eration with third-generation chemotherapy; but this PS of 0 or 1. All dose levels were tolerable for regi-
may be the only phase III trial to attempt to address men 1 (n = 18: 15 NSCLC and three esophageal
the platinum question, which, to be frank, arises cancers) and regimen 2 (n = 12: all NSCLC). In
constantly. Regardless, a platinum backbone grafted regimen 1, one dose-limiting toxicity consisting of
onto XRT is the standard for combination therapy in grade 4 esophagitis/anorexia occurred at 500 mg/m2.
LA-NSCLC. Whether newer agents such as peme- Grade 3 neutropenia was the main hematologic tox-
trexed or abraxane can do as well or better than icity and occurred in three of 18 patients. In regimen
established agents in LA-NSCLC remains to be seen. 2, one dose-limiting toxicity consisting of grade 3
Focus on Pemetrexed and Nab-Paclitaxel 767

esophagitis occurred at a pemetrexed dose of Choy et al. (2010) have adopted a similar regimen
500 mg/m2 and carboplatin dose of AUC = 6. Grade looking at combination pemetrexed with either cis-
3/4 leukopenia was the main hematologic toxicity and platin or carboplatin in combination with XRT.
occurred in four of 12 patients. Four complete Patients with inoperable stage IIIA/B NSCLC were
responses (two pathologically proven) and eight partial randomized to pemetrexed 500 mg/m2 every 21 days
responses were observed in those receiving regimen 2. combined with carboplatin AUC = 5 (PCb) or cis-
When systemically active chemotherapy doses were platin 75 mg/m2 (PC) intravenously every 21 days for
reached, further dose escalation was discontinued, and 3 cycles. All points received concurrent RT 64–68 Gy
a phase II dose-range was established: pemetrexed (2 Gy/day, 5 days/week, days 1–45). Consolidation
500 mg/m2 in combination with carboplatin pemetrexed 500 mg/m2 IV every 21 days for 3 cycles
AUC = 5–6. The authors concluded that this regimen began 3 weeks after completion of chemoradiation.
was feasible and have since exported it to the coop- The primary endpoint of this ongoing trial was 2-year
erative groups. Of note, in contrast to many other overall survival; secondary endpoints included tox-
regimens, systemically active chemotherapy doses icity, response rate, time to progression, and median
of pemetrexed and carboplatin could be given survival. Since June 2007, 72 patients have been
concurrently with XRT, without untoward toxicity. evaluated: 34 received PCb: 38 received PC. Average
To further determine efficacy, safety, and dose compliance was 89% or higher for each agent.
optimal dosing, the cancer and leukemia Group B Average dose compliance for CRT was 91.2% for
study 30407 recently completed evaluating this those receiving PCb and 85.5% for those receiving
regimen in patients with unresectable stage III PC. Dose interruptions occurred in 23 patients.
NSCLC. Govindan et al. (2009) mounted a ran- Amongst 15 patients evaluable for response in the
domized phase II trial of pemetrexed and carboplatin PCb arm, there was one CR (6.7%) and six patients
in combination with radical thoracic radition with PR (40.0%); seven patients (46.7%) had stable
(70 Gy). Half the enrollees also received cetuximab disease and one had PD (6.7%). The PC arm featured
[C225]. In addition to two full cycles of carboplatin 20 evaluable patients, of whom one (5.0%) had a CR,
and pemetrexed during XRT and two administered 11 (55%) had a PR, five (25.0%) had SD, and three
after completion of XRT, all patients were assigned (15.0%) had PD. The authors of this abstract justifi-
to receive four additional cycles of single-agent ably concluded that pemetrexed and radical thoracic
pemetrexed 500 mg/m2 as consolidation. Due to the RT in combination with either carboplatin or cisplatin
timeframe in which the trial was designed and appeared well-tolerated in the treatment of locally
conducted, all histologies were allowed. Although advanced NSCLC.
the overall incidence of grade 3/4 toxicities was Other studies have evaluated pemetrexed and tho-
fairly high, they were generally tolerable and racic RT exclusively in combination with cisplatin. A
essentially similar in both arms, with the exception study led by Brade et al. (2011) was restricted to good
of skin rash: 4% in the non-cetuximab arm versus prognosis patients with \5% weight loss and good
41% in the cetuximab arm. There was no obvious performance status. Enrollees received two cycles of
exacerbation of esophagitis or pulmonary toxicity pemetrexed (300, 400, or 500 mg/m2 on days 1 and 22
compared to other similar trials incorporating plati- for dose Levels 1, 2, and 3/4, respectively) and cis-
num combinations. Median survival proved to be platin (25 mg/m2 days 1–3 for dose Levels 1–3;
22 months in both arms. Because data in patients 20 mg/m2 days 1–5 for dose Level 4) concurrent with
with advanced NSCLC showed a survival benefit for thoracic radiation (61–66 Gy in 31–35 fractions).
pemetrexed predominantly in nonsquamous NSCLC Consolidation consisted of two cycles of pemetrexed/
(Scagliotti et al. 2008), the results were analyzed by cisplatin (500 mg/m2, 75 mg/m2) 21 days apart, after
histology. For squamous versus nonsquamous his- concurrent therapy. Between January 2006 and
tology, median OS was 18 months and 22 months October 2007, 16 patients were accrued. No dose-
and 18-month OS proved 48 and 56%, respectively. limiting toxicities were observed. Median radiation
These differences were not statistically significant, dose was 64 Gy (range, 45–66 Gy). Rates of signifi-
but suggest a potential preferential advantage for cant Grade 3/4 hematologic toxicity were 38 and 7%,
pemetrexed in non-squamous histology. respectively. One patient experienced Grade 3 acute
768 C. J. Langer

esophagitis, and two experienced late grade 3 esoph- followed by concurrent radiation and pemetrexed for
ageal strictures, successfully managed with dilatation. 2 cycles or to concurrent radiation and pemetrexed
One patient experienced Grade 3 pneumonitis. The for 2 cycles, followed by three cycles of consolida-
overall response rate was 88%. At median follow-up of tion cisplatin-pemetrexed (ClinicalTrials.gov. NCT0
17.2 months, 1-year overall survival was 81%. Long- 0497315). A separate trial orchestrated by the
term PFS and OS are still pending. The authors con- Intergroupe Francophone de Cancerologie Thoraci-
cluded that full-dose systemic pemetrexed was safe in que assesses concurrent pemetrexed 500 mg/m2 and
combination with full-dose cisplatin and thoracic cisplatin 75 mg/m2 IV every 3 weeks for 4 cycles in
radiation in Stage IIIA/B NSCLC, and that pemetrexed combination with C225 and full dose RT (66 Gy;
was, in fact, the first third-generation cytotoxic agent 2 Gy/fx) (ClinicalTrials.gov. NCT01102231). Eligi-
tolerable at full dose in this setting. Phase II study bility stipulates stage III, non-squamous cell NSCLC
evaluating dose Level 4 is ongoing. and performance status 0–1. An aborted trial con-
Another ongoing, global, randomized Phase III ducted the Hoosier Oncology Group grafted single-
trial titled PROCLAIM [H3E-MC-JMIG] in the agent pemetrexed onto standard dose thoracic RT
context of chemoradiation compares pemetrexed– (60 Gy) in poor prognosis patients with compro-
cisplatin followed by single-agent pemetrexed during mised PS or >10% baseline weight loss. (Clinical-
the consolidation period with standard chemoradia- Trials.gov. NCT00497315. Unfortunately, only eight
tion (ClinicalTrials.gov. NCT00686959; H3E-MC- patients were enrolled between 8/08 and 12/10
JMIG). Eligibilty stipulates fit patients with stage leading to discontinuation of this study. Finally,
IIIA/B non-squamous, non-small cell lung cancer there is some suggestion that pemetrexed may help
patients. Enrollees are allowed up to 10% weight reduce the incidence of CNS metastases. The Line-
loss from baseline. Patients in the investigational berger Cancer Center of UNC, led by Stinchcombe
arm receive pemetrexed 500 mg/m2 plus cisplatin and colleagues is assessing concurrent pemetrexed
75 mg/m2 every three weeks for 3 cycles during and WBRT in NSCLC patients with newly diag-
concurrent thoracic radiation to 66 Gy followed by nosed brain metastases (ClinicalTrials.gov. NCT00
consolidation with pemetrexed 500 mg/m2 for 4 280748 UNC-LCCC 0409). Thirty patients with KPS
cycles. In the standard arm, patients receive cisplatin of 70 or greater were accrued between 5/05 and
and etoposide for 2 cycles during concurrent thoracic 6/09. The results are not yet available.
radiation followed by two additional cycles of con-
solidation, with clinician’s choice of either cisplatin/
etoposide, cisplatin/vinorelbine, or carboplatin/pac- 3 Abraxane
litaxel. The primary endpoint is overall survival;
accrual of 900 patients is planned (ClinicalTrials. Abraxane [nab-paclitaxel] or ABI007 is a novel,
gov. NCT00686959; H3E-MC-JMIG). As of 2/28/11, solvent-free 130 nm nanoparticle albumin-bound
316 individuals have been enrolled. To date, no formulation of paclitaxel, designed without the dose-
untoward or unusual toxicities have been noted. The limiting solvent Cremophor EL used in the standard
role of pemetrexed in LA-NSCLC will likely be solvent-based paclitaxel. Like paclitaxel, it is also
determined by the results of this trial. If it is formally a radiosensitizer. A dose-finding study showed a
approved in this setting based on survival, there higher maximum tolerated dose for nab-paclitaxel
is little doubt that this agent will supplant etopo- compared to solvent-based paclitaxel and linear
side in the therapy of locally advanced, unresectable pharmacokinetics (Ibrahim et al. 2002; Nyman et al.
NSCLC. 2005). Nab-paclitaxel is well tolerated without ste-
Meanwhile, ClinicalTrials.gov. lists at least ten roid or H1/H2 blocker premedication when given at
ongoing studies evaluating pemtrexed in combina- doses higher than standard solvent-based paclitaxel;
tion with radiation in locally advanced NSCLC, as it has produced significant tumor response in patients
well as other settings. Of note, a randomized phase with non-small cell lung cancer (Green et al. 2006),
II sequencing trial, being conducted in Amsterdam, metastatic breast cancer (Ibrahim et al. 2005), and
assigns ‘‘good prognosis’’ patients to either induction head and neck and anal canal cancers (Damascelli
therapy with pemetrexed–cisplatin for 3 cycles et al. 2001).
Focus on Pemetrexed and Nab-Paclitaxel 769

Preclinical xenograft studies have shown that cel- (82 vs. 65 mg/m2/wk). With respect to overall
lular transport of nab-paclitaxel differs from that of response rate (ORR), nab–PC was superior to PC both
standard solvent-based paclitaxel. Albumin is trans- by independent radiographic review [33 vs. 25%,
ported via a gp60-mediated mechanism into endo- P = 0.005; 1.31 response ratio (RR), 95% CI: 1.08,
thelial cells and via a secreted protein acidic and rich 1.59], and by investigator review [37 vs. 30%,
in cysteine (SPARC)–mediated mechanism into P = 0.008; 1.26 RR, CI: 1.06, 1.50]. Histologic
SPARC-overexpressing tumor cells, resulting in analysis showed significantly improved ORR for nab–
selective nab-paclitaxel accumulation in tumors PC (n = 228) versus PC (n = 221) in squamous cell
(Desai et al. 2006; Sparreboom et al. 2005; ABI 007. carcinoma (SQC) patients (41 vs. 24%, P \ 0.001,
Drugs 2004) with a 33% higher intra-tumoral con- IRR; 1.67 RR). Nab–PC was well tolerated, with
centration of paclitaxel following administration of significantly improved safety profile, including
ABI-007compared with an equal dose of cremophor- reduced neurotoxicity and hematologic toxicity ver-
based paclitaxel in the MX-1 xenograft model sus. PC. Longterm PFS and OS data are still pending,
(Sparreboom et al. 2005). In addition, elevated but this phase III trial has rekindled interest in the use
expression of Cav-1 has been identified in various of abraxane concurrently with radiation, although
tumor types including breast, prostate, lung, pancre- there remains a relative paucity of trials investigating
atic, renal, and esophageal cancer. Caveolin-1 over- this strategy.
expression has been linked to tumor aggressiveness In preclinical models, such as murine ovarian
and poor patient prognosis. Albumin-bound nano- carcinoma [OCa-1], abraxane results in radio-sensi-
particles (e.g. Abraxane) may help exploit Caveolin-1 tization of the tumor without increased normal tissue
to deliver more drug selectively to tumors. The damage (Wiedenmann et al. 2007). In one study, mice
unique formulation also enables paclitaxel to traverse bearing syngeneic ovarian or mammary carcinomas
the circulation more seamlessly than standard paclit- were treated with nab-paclitaxel, radiation, or a
axel wedded to a cremaphor vehicle. As a conse- combination of both. Nab-paclitaxel was administered
quence, nano-particle technology can help reduce the at 90 mg/kg, 1.5 times the maximum tolerated dose
toxicities typically associated with paclitaxel, for solvent-based paclitaxel. Endpoints included
including neuropathy, myalgias, and arthralgias. antitumor efficacy (growth delay, radiocurability, and
Abraxane was first approved in metastatic and cellular effects) and normal tissue toxicity (gut and
recurrent breast cancer on the basis of a phase III trial skin). Nab-paclitaxel demonstrated single-agent anti-
comparing this agent to standard paclitaxel (Gradishar tumor efficacy against both tumor types and acted as a
et al. 2005). Abraxane achieved a superior overall radiosensitizer. Combined with radiation, nab-paclit-
response rate and a significantly longer time to tumor axel produced supra-additive effects when given
progression (P = 0.029). The incidence of peripheral before radiation. Nab-paclitaxel significantly increased
neuropathy was somewhat higher, but easily managed radiocurability by reducing the dose yielding 50%
and more rapidly reversible, compared to standard tumor cure (TCD50) from 54.3 to 35.2 Gy. Tumor
paclitaxel. There was also a lower incidence of neu- histology following nab-paclitaxel treatment was
tropenia. The much shorter infusion schedule also characterized by pronounced necrotic and apoptotic
favored abraxane over paclitaxel [10–30 min com- cell death and mitotic arrest. Nab-paclitaxel did not
pared to 3 h]. increase the normal tissue radio-response. These
A recent phase III trial enrolling over 1,000 improved effects were achieved without increased
patients with advanced NSCLC compared weekly normal tissue toxicity to either rapidly or slowly
abraxane in combination with carboplatin (nab–PC) proliferating normal tissues, although the drug dose
administered every 3 weeks to standard paclitaxel and was 50% higher than the maximum tolerated dose of
carboplatin (PC) every 3 weeks; the results showed solvent-based paclitaxel.
therapeutic equivalency between the two regimens Work by Harari et al. (2010) has demonstrated
and improved response rate in squamous cell carci- similar promise. Optical imaging using phase display
noma (Socinski et al. 2010). Baseline and histologic technology showed that HVGGSSV-guided nab-
characteristics were well balanced. Dose intensity of paclitaxel selectively targeted irradiated tumors and
paclitaxel was higher in nab–PC compared to PC showed 1.48 ± 1.66 photons/s/cm2/sr greater radiance
770 C. J. Langer

compared with SGVSGHV-nab-paclitaxel, and are considered poor risk based either on NCI CTC
1.49 ± 1.36 photons/s/cm2/sr greater than nab-paclit- performance status (PS) 2 or C10% weight loss
axel alone (P \ 0.05). Bio-distribution studies showed within the past 3 months.
a more than fivefold increase in paclitaxel levels
within irradiated tumors in HVGGSSV-nab-paclitaxel
treated groups compared with either nab-paclitaxel 4 Conclusions
or SGVSGHV-nab-paclitaxel at 72 h. Both Lewis
lung carcinoma and H460 lung carcinoma murine Pemetrexed has established its role in phase II and
models showed significant tumor growth delay for phase III trials in LA-NSCLC. It is clear, at this point,
HVGGSSV-nab-paclitaxel compared with nab-paclit- that this agent can be combined at full systemic
axel, SGVSGHV-nab-paclitaxel,and saline controls. dose with either carboplatin or cisplatin with
HVGGSSV-nab-paclitaxel treatment induced a sig- respectable PFS and OS in cooperative group studies
nificantly greater loss in vasculature in irradiated in LA-NSCLC and acceptable toxicity. Whether
tumors compared with unirradiated tumors, nab- pemetrexed and platinum can supplant etoposide
paclitaxel, SGVSGHV-nab-paclitaxel, and untreated and cisplatin will be determined by the results of
controls. an ongoing phase III trial that is enrolling well.
In the clinical realm, an ongoing phase I/II trial in Nab-paclitaxel, while clearly radiosensitizing in pre-
LA-NSCLC at Vanderbilt-Ingraham Cancer Center clinical and xenograft models, is much further behind
(ClinicalTrials.gov. NCT00544648), is evaluating in clinical trials integrating this agent with XRT in
carboplatin (AUC 2) IV over 30 min following LA-NSCLC. Assuming the results of phase III testing
nab-paclitaxel, once/week 9 7 weeks, in combina- in advanced NSCLC leads to its approval, then interest
tion with 3D conformal radiotherapy or intensity- in investigating this agent in LA-NSCLC will grow.
modulated radiation therapy (IMRT), 2.0 Gy/day 9
5 days/week for 33 days during weeks 1–7 (total
dose = 66 Gy). The starting dose for nab-paclitaxel References
is 40 mg/m2. The primary endpoint is determination
of the maximum tolerated dose (MTD) of paclitaxel ABI 007. Drugs RD (2004) 5:155–159 (For more details see
albumin-stabilized nanoparticle formulation when doi: 10.1200/JCO.2005.03.7135
given concurrently with carboplatin and radiation Aupérin A, Rolland E, Curran WJ et al (2007) Concomitant
followed by two courses of paclitaxel albumin- radio-chemotherapy (RT-CT) versus sequential RT–CT in
locally advanced non-small cell lung cancer (NSCLC): a
stabilized nanoparticle formulation with carboplatin meta-analysis using individual patient data (IPD) from
as consolidation. Once the MTD is established, the randomised clinical trials (RCTs): A1-04. J Thorac Oncol
phase II component of the study will determine 2:S310
progression-free survival in patients with stage III Brade A, Bezjak A, MacRae R (2011) Phase I trial of radiation
with concurrent and consolidation pemetrexed and cisplatin
unresectable non-small cell lung cancer treated. in patients with unresectable stage IIIA/B non-small cell
Secondary objectives focus on overall survival as well lung cancer. Int J Radiat Oncol Biol Phys 79(5):1395–1401
as biocorrelatives including tumor specimen analysis [Epub 2010 Jun 3]
for the secreted protein acidic and rich in cysteine Choy H, Curran WJ, Scott CB , Bonomi P, Travis P, Haluschak
J, Belani CP (2002) Preliminary report of locally advanced
(SPARC) gene expression. multimodality protocol (LAMP): ACR 427: a randomized
A separate trial in poor prognosis patients with phase II study of three chemo-radiation regimens with
locally advanced NSCLC will evaluate induction paclitaxel, carboplatin, and thoracic radiation (TRT) for
therapy with carboplatin and weekly nab-paclitaxel, patients with locally advanced non small cell lung cancer
(LA-NSCLC). Proc Am Soc Clin Oncol 21
followed by concurrent full dose radiation and erl- Choy H, Schwartzberg LS, Dakhil SR (2010) Ongoing phase II
otinib (ClinicalTrials.gov. NCT00553462). While this study of pemetrexed plus carboplatin or cisplatin with
study does not assess the radiosensitizing properties concurrent radiation therapy followed by pemetrexed con-
of nab-paclitaxel directly, it attempts to mitigate solidation in patients with favorable-prognosis inoperable
stage IIIA/b non-small cell lung cancer: Interim update.
toxicity by incorporating this agent in the induction J Clin Oncol 28:15s [suppl; abstr 7082]
setting prior to definitive RT in patients who might Ciuleanu T, Brodowicz T, Zielinski C, Kim JH, Krzakowski M,
have trouble tolerating standard treatment. Enrollees Laack E, Wu YL, Bover I, Begbie S, Tzekova V, Cucevic B,
Focus on Pemetrexed and Nab-Paclitaxel 771

Pereira JR, Yang SH, Madhavan J, Sugarman KP, Peterson Hariri G, Yan H, Wang H, Han Z, Hallahan DE (2010)
P, John WJ, Krejcy K, Belani CP (2009) Maintenance Radiation-guided drug delivery to mouse models of lung
pemetrexed plus best supportive care versus placebo plus cancer. Clin Cancer Res 16(20):4968–4977 [Epub 2010 Aug
best supportive care for non-small cell lung cancer: a 27]
randomised, double-blind, phase 3 study. Lancet Ibrahim NK, Desai N, Legha S et al (2002) Phase I and
374(9699):1432–1440 [Epub 2009 Sep 18. PMID: pharmacokinetic study of ABI-007, a Cremophor-free,
19767093] protein-stabilized, nanoparticle formulation of paclitaxel.
Curran W, Paulus R, Langer CJ, Komaki R, Lee JS, Hauser S, Clin Cancer Res 8:1038–1044
Movsas B, Wasserman T, Russell A, Byhardt R, Machtay Ibrahim NK, Samuels B, Page R et al (2005) Multicenter phase
M, Sause W, Cox JD (2010) Phase III Comparison of II trial of ABI-007, an albumin-bound paclitaxel in women
sequential vs concurrent chemo-radiation for patients with with metastatic breast cancer. J Clin Oncol 23:6019–6026
unresected stage III non-small cell lung cancer (NSCLC): Movsas B, Scott C, Langer C et al (2005) Randomized trial of
report of radiation therapy oncology group (RTOG) 9410. amifostine in locally advanced non-small cell lung cancer
JNCI (in press) patients receiving chemotherapy and hyperfractionated
Damascelli B, Cantu G, Mattavelli F et al (2001) Intraarterial radiation: radiation therapy oncology group trial 98-01.
chemotherapy with polyoxyethylated castor oil free paclit- J Clin Oncol 23(10):2145–2154
axel, incorporated in albumin nanoparticles (ABI-007): Nyman DW, Campbell KJ, Hersh E et al (2005) Phase I and
phase II study of patients with squamous cell carcinoma of pharmacokinetics trial of ABI-007, a novel nanoparticle
the head and neck and anal canal: preliminary evidence of formulation of paclitaxel in patients with advanced nonhe-
clinical activity. Cancer 92:2592–2602 matologic malignancies. J Clin Oncol 23:7785–7793
Desai N, Trieu V, Yao Z et al (2006) Increased antitumor Scagliotti GV, Parikh P, von Pawel J et al (2008) Phase III
activity, intratumor paclitaxel concentrations, and endothe- study comparing cisplatin plus gemcitabine with cisplatin
lial cell transport of cremophor-free, albuminbound paclit- plus pemetrexed in chemotherapy-naive patients with
axel, ABI-007, compared with cremophor-based paclitaxel. advanced-stage non-small cell lung cancer. J Clin Oncol
Clin Cancer Res 12:1317–1324 26:3543–3551
Furuse K et al (1999) Phase III study of concurrent versus Seiwert TY, Connell PP, Mauer AM et al (2007) A phase I
sequential thoracic radiotherapy in combination with mito- study of pemetrexed, carboplatin, and concurrent radiother-
mycin, vindesine, and cisplatin in unresectable stage III apy in patients with locally advanced or metastatic non-
non-small cell lung cancer. J Clin Oncol 17(9):2692–2699 small cell lung or esophageal cancer. Clin Cancer Res
Green MR, Manikhas GM, Orlov S et al (2006) Abraxane(R), a 13:515–522
novel Cremophor(R)-free, albumin-bound particle form of Socinski MA, Bondarenko IN, Karaseva NA (2010) Results of
paclitaxel for the treatment of advanced non-small cell lung a randomized, phase III trial of nab-paclitaxel (nab-P) and
cancer. Ann Oncol 17:126–138 carboplatin (C) compared with cremophor-based paclitaxel
Govindan R, Bogart J, Wang X et al (2009) Phase II study of (P) and carboplatin as first-line therapy in advanced non-
pemetrexed, carboplatin, and thoracic radiation with or small cell lung cancer (NSCLC). J Clin Oncol 28:18s
without cetuximab in patients with locally advanced [suppl; abstr LBA7511]
unresectable non-small cell lung cancer: CALGB 30407. Sparreboom A, Baker SD, Verweij J (2005) Paclitaxel repack-
Program and abstracts of the 45th annual meeting of the aged in an albumin-stabilized nanoparticle: handy or just a
American Society of Clinical Oncology, May 29–June 2, dandy? J Clin Oncol 23:7765–7767
Orlando, Florida, Abstract 7505 Wiedenmann N, Valdecanas D, Hunter N et al (2007)
Gradishar WJ, Tjulandin S, Davidson N et al (2005) Phase III Radiocurability and therapeutic gain 130-nm albumin-
trial of nanoparticle albumin-bound paclitaxel compare with bound paclitaxel enhances tumor. Clin Cancer Res 13:
polyethylated castor oil-based paclitaxel in women with 1868–1874
breast cancer. J Clin Oncol 23:7794–7803 Yamamoto N, Nakagawa K, Nishimura Y, Tsujino K, Satouchi
Hanna N, Shepherd FA, Fossella FV, Pereira JR, De Marinis F, M, Kudo S, Hida T, Kawahara M, Takeda K, Katakami N,
von Pawel J, Gatzemeier U, Tsao TC, Pless M, Muller T, Sawa T, Yokota S, Seto T, Imamura F, Saka H, Iwamoto Y,
Lim HL, Desch C, Szondy K, Gervais R, Shaharyar, Semba H, Chiba Y, Uejima H, Fukuoka M (2010) Phase III
Manegold C, Paul S, Paoletti P, Einhorn L, Bunn PA Jr study comparing second- and third-generation regimens
(2004) Randomized phase III trial of pemetrexed versus with concurrent thoracic radiotherapy in patients with
docetaxel in patients with non-small cell lung cancer unresectable stage III non-small cell lung cancer: West
previously treated with chemotherapy. J Clin Oncol Japan Thoracic Oncology Group WJTOG0105. J Clin Oncol
22(9):1589–1597 28(23):3739–3745 Epub 2010 Jul 12
Novel Targeted Agents
and Radiopharmaceuticals in Lung Cancer
Martin J. Edelman and Nadia Ijaz

Contents Abstract
The use of chemotherapy in addition to radiation
1 Overview ................................................................... 773 has unquestionably improved outcomes in both
2 Epidermal Growth Factor Receptor ..................... 775 small cell and non-small cell lung cancer.
2.1 EGFR Monoclonal Antibodies.................................. 775 However, as in advanced disease, a plateau in
2.2 EGFR Tyrosine Kinase Inhibitors ............................ 777 efficacy has been reached. Over the past several
3 Vascular Endothelial Growth Factor.................... 778 decades numerous receptors and pathways respon-
3.1 VEGF Antibodies ...................................................... 778 sible for normal cellular growth, maintenance and
3.2 VEGFR TK Inhibitors ............................................... 779 development have been identified. Derangements of
4 PARP Inhibitors ...................................................... 780 these pathways frequently result in cancer, perpet-
5 Proteasome Inhibitors ............................................. 780 uate cancer or result in resistance to treatment with
chemotherapy and/or radiation. New agents target-
6 HDAC Inhibitors ..................................................... 781
ing these pathways are now becoming available.
7 mTOR Inhibitors ..................................................... 782 Many of these have the potential to potentiate
8 IGF-IR Inhibitors .................................................... 782 existing agents and/or radiation. In addition,
radiation can itself be targeted by coupling radio-
9 Cyclooxygenase-2 Inhibitors................................... 783
isotopes to a moiety that will bind with receptors
10 ALK Targeted Inhibitors........................................ 784 that are aberrantly expressed in cancer.
11 Aurora Kinase and Polo-like
Kinase Inhibitors ..................................................... 784
12 Hedgehog Pathway Inhibitors................................ 785
13 Radiopharmaceuticals ............................................. 785
1 Overview
References.......................................................................... 787 Many commonly employed antineoplastic agents
including the platinums, vinca alkaloids, and taxanes
have been incorporated into chemoradiotherapy reg-
imens that have improved the potential for cure in
patients with locally advanced (stages IIIa, b) non-
small cell lung cancer (NSCLC). Platinum and eto-
poside combined with radiotherapy is the standard,
and potentially curative, approach for limited stage
M. J. Edelman (&)  N. Ijaz (stages I–IIIB) small cell lung cancer. The extensive
University of Maryland Greenebaum Cancer Center,
research done along the lines of molecular structures
Rm. N9E08 22 S. Greene Street, Baltimore,
MD 21201, USA and biochemical pathways over the past several dec-
e-mail: medelman@umm.edu ades has resulted in numerous new agents with

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 773
DOI: 10.1007/174_2011_312, Ó Springer-Verlag Berlin Heidelberg 2011
774 M. J. Edelman and N. Ijaz

Table 1 Novel targets and Class Agent(s)


agents currently under clinical
investigation VEGF Bevacizumaba
Sorafeniba
Sunitiniba
Vandetaniba
Cediranib
Epidermal growth factor receptor (EGFR) Cetuximab (C225)a
Panitumumaba
Erlotiniba (OSI 774)
Gefitiniba (ZD 1839)
PARP inhibitor Veliparib (ABT-888)
Iniparib (SAR240550, BSI-201)
Olaparib
PF1367338 (AG-014699)
MK 4827
AZD2281 (KU-0054436)
Proteasome inhibitors Bortezomiba
Carfilzomib
MLN-9708
CEP-18770
HDAC inhibitors Vorinostata
Etinostat
COX-2 Celecoxiba
Apricoxib
ALK inhibitor Crizotinib (PF1066)
IGF-R inhibitors Figitumumab
IMC-A12
RI-507
AMG-479
SCH-717454
INSM-18
OSI-906
XL-228
Hedgehog pathway inhibitors GDC-0449
PF-04449913
TAK-441
IPI-926
LDE225
mTOR inhibitors Sirolimusa
Everolimusa
Radaforolimus
(continued)
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 775

Table 1 (continued) Class Agent(s)


Polo-like kinase inhibitors BI 6727 (Volasertib)
TAK 960
NMS-1286937
Aurora kinase inhibitors AT9283
AS703569
CYC116
PF-03814735
MLN8237
LBH589B
SNS 314
AMG900
GSK1070916A
a
FDA approved for at least one indication

potential benefit for the treatment of cancer. These EGFR, activating EGFR mutations were discovered
‘‘targeted’’ drugs act through pathways necessary for and found to occur in approximately 10% of patients
the development and continued growth of malignan- with NSCLC. These mutations are both prognostic
cies. Many of them have the potential for combination and predictive of response to EGFR tyrosine kinase
with existing agents as well as with radiation. Some inhibitors (TKIs) (Lynch et al. 2004; Paez et al.
of these agents are still in the early phase of devel- 2004). Two major families of agents, antibodies, and
opment while others are in standard clinical use. None TKIs, have been developed that target the EGFR.
has yet to be conclusively demonstrated to be of use Two antibodies are approved that target the external
in combination with radiotherapy in lung cancer, domain (cetuximab, panitumimab) and two TKIs
Table 1. (gefitinib and erlotinib) target the ATP binding site of
This chapter will discuss some of the many novel the internal domain.
agents and pathways that are currently under inves-
tigation in NSCLC. It will focus on the potential of
these agents to be employed with radiotherapy. 2.1 EGFR Monoclonal Antibodies
Targeted radiopharmaceuticals are also discussed.
In many cases, these agents are just entering clinical The monoclonal antibodies (MAbs) are directed at the
trials and information is limited. Ongoing studies are extra-cellular domain of the EGFR receptor and
listed with their National Clinical Trials number include drugs such as cetuximab (C225), pani-
(clinicaltrials.gov) to allow the reader to determine tumumab and matuzumab (EMD 72000). Cetuximab,
the status of the study. a chimerized antibody of the IgG1 subclass, was
originally derived from a mouse myeloma cell line.
The chimerization process resulted in an antibody
2 Epidermal Growth Factor Receptor with a relative affinity five-fold greater than the
murine monoclonal antibody (Mendelsohn et al.
Constitutive activation of the epidermal growth factor 1990). Cetuximab blocks binding of EGF and TGFa
receptor (EGFR) may be a critical step in malignant to EGFR and inhibits ligand-induced activation of this
proliferation, angiogenesis, and metastasis (Wood- tyrosine kinase receptor. Cetuximab also stimulates
burn 1999). The initial impetus to target the EGFR EGFR internalization, effectively removing the
was based upon its frequent overexpression in many receptor from the cell surface for interaction with
malignancies including NSCLC and the association of ligand (Baselga et al. 1993). Cetuximab binds spe-
overexpression with poor prognosis. As a conse- cifically to the epidermal growth factor receptor
quence of the development of agents targeting the (EGFR, HER1, c ErbB-1) on both normal and tumor
776 M. J. Edelman and N. Ijaz

cells, and competitively inhibits the binding of EGF the trial failed to identify any predictive biomarkers
and other ligands, such as TGFa. Binding of cetux- (Khambata-Ford et al. 2010). In contrast, the FLEX
imab to the EGFR blocks phosphorylation and acti- trial combined cisplatin, vinorelbine, and cetuximab
vation of receptor-associated kinases, resulting in versus cetuximab alone in a population of advanced
inhibition of cell growth, induction of apoptosis, and lung cancer patients selected for EGFR expression by
decreased matrix metalloproteinase and vascular immunohistochemistry (though the degree of expres-
endothelial growth factor (VEGF) production. sion was not specified) and found that the EGFR
Cetuximab has shown to enhance the activity of inhibitor resulted in a significantly improved overall
cytotoxic drugs in colorectal cancer and radiotherapy survival (Pirker et al. 2009). One thousand one hun-
in head and neck cancer. Single agent activity has also dred and twenty five patients were randomly assigned
been demonstrated in colorectal cancer. Cetuximab is to chemotherapy plus cetuximab (n = 557) or che-
approved for the treatment of colorectal cancer also motherapy alone (n = 568). The experimental arm
approved in combination with radiotherapy for the demonstrated superior survival (median 11.3 vs.
treatment of head and neck cancer. Panitumumab is a 10.1 months; HR = 0.871; P = 0.044). Recently pre-
high-affinity fully human IGG2 monoclonal antibody sented data indicate that the benefit see with cetuximab
against EGFR. Compared with cetuximab, it is much is dependent upon the degree of EGFR experiment
less likely to cause severe infusion reactions, does not (Pirker et al. 2011).
require premedications nor a loading dose. It is cur- None of the targeted agents has been evaluated as
rently approved as a single agent in advanced colo- thoroughly with radiotherapy as cetuximab. A phase
rectal cancer in Kras wild type disease. III trial in head and neck cancer has demonstrated
Two phase III trials of cetuximab in combination superiority of cetuximab combined with radiotherapy
with chemotherapy have been performed in NSCLC versus radiotherapy alone and has led to its approval
with variable results. In the United States, a random- in combination with radiotherapy (Bonner et al.
ized multicenter phase III trial BMS099 evaluated 2006). The efficacy and safety of cetuximab were
Cetuximab and first-line taxane/carboplatin chemo- studied in combination with radiation therapy in a
therapy as first-line treatment in advanced NSCLC randomized, controlled trial of 424 patients with
(Lynch et al. 2010). Six hundred and seventy six locally or regionally advanced squamous cell carci-
chemotherapy-naïve patients with advanced stage noma of the head and neck versus radiation alone.
NSCLC were enrolled. There was no requirement to Four hundred twenty-four patients with Stage III/IV
demonstrate EGFR expression. Patients were ran- SCCHN of the oropharynx, hypopharynx, or larynx
domly assigned to cetuximab and taxane/carboplatin with no prior therapy were randomized (1:1) to
versus taxane/carboplatin. The taxane was at the receive cetuximab plus radiation therapy (211
investigator’s discretion and could be either paclitaxel patients) or radiation therapy alone (213 patients).
or docetaxel. The primary end point was progression- Radiation therapy was administered for 6–7 weeks as
free survival assessed by independent radiologic once-daily, twice-daily, or concomitant boost. Start-
review committee. The median progression-free sur- ing one week before radiation, cetuximab was
vival (as determined by the IRRC) was 4.40 months administered as a 400-mg/m2 initial dose, followed by
with cetuximab/TC versus 4.24 months with TC 250 mg/m2 weekly for the duration of radiation
(hazard ratio [HR] = 0.902; 95% CI, 0.761 to 1.069; therapy (6–7 weeks). Cetuximab was administered
P = 0.236). Median overall survival was 9.69 months 1 h prior to radiation therapy, beginning week 2.
with cetuximab/TC versus 8.38 months with TC The patient characteristics were similar across the
(HR = 0.890; 95% CI, 0.754 to 1.051; P = 0.169). study arms. 56% of the patients received radiation
ORR-IRRC was 25.7% with cetuximab/TC versus therapy with concomitant boost, 26% received once-
17.2% with TC (P = 0.007). The safety profile of this daily regimen, and 18% twice-daily regimen. The
combination was manageable and consistent with its main outcome measure of this trial was duration of
individual components. The addition of cetuximab to locoregional control, which was superior for the
TC did not significantly improve the primary end combination of cetuximab with radiation (24.4 vs.
point, though there was significant improvement in the 14.0 mo, HR = 0.68, P = 0.005) with an advantage
overall response rate. An analysis of specimens from also demonstrated for the secondary endpoint of
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 777

overall survival (49.0 vs. 29.3 mo, HR = 0.74, contrast to the lack of benefit from the addition of
P = 0.03). EGFR TKIs to cytotoxics. Ultimately, approval for
Two major studies have been reported that evalu- erlotinib resulted from the BR.21 trial conducted by
ate cetuximab in combination with chemoradiother- the National Cancer Institute of Canada Clinical
apy in locally advanced NSCLC, with conflicting Trials Group which compared erlotinib to placebo in
results. RTOG 0324 (n = 93) combined carboplatin, previously treated patients with NSCLC (Shepherd
taxol, radiotherapy, and cetuximab in treatment of et al. 2005). The results of this study led to approval
inoperable stage III patients. There was no selection of erlotinib for the treatment of NSCLC after pro-
based upon EGFR status. The median survival of gression on 1 or 2 prior chemotherapy regimens. This
22.7 months is among the best ever demonstrated for trial demonstrated improved response rate, time to
a cooperative group study population. Importantly, progression, survival, and quality of life compared
other than rash, there was little evidence that the use with placebo. A similar, larger trial termed, Iressa
of cetuximab was associated with increased toxicity Survival Evaluation in Lung Cancer (ISEL) compared
(Blumenschein et al. 2011). Another cooperative gefitinib with placebo. This study produced similar
group study, CALGB 30407, evaluated carboplatin, results in terms of response and quality of life; how-
pemetrexed, cetuximab, and concurrent radiotherapy ever, the 1-year survival endpoint failed to reach the
versus carboplatin, pemetrexed, and concurrent statistically defined criteria for superior survival and
radiotherapy. Similar to the RTOG trial, other than resulted in withdrawal of approval in the United
increased skin rash, there was no difference in tox- States, though the drug is still approved in many parts
icity. However, survival on the two arms was similar of Europe and Asia. Subsequent studies have evalu-
and the addition of cetuximab was not felt to be ated gefitinib versus carboplatin/paclitaxel in first-line
beneficial (Govindan et al. 2009). Currently, a ran- therapy and against docetaxel in second line therapy
domized phase III trial is in progress (RTOG 0617, and found the agent to be active and well tolerated.
NCT00533949) is testing the addition of cetuximab as Erlotinib is also approved for use in the maintenance
well as radiation dose to chemoradiotherapy in locally setting after initial benefit from platinum based che-
advanced, inoperable NSCLC. motherapy (Cappuzzo et al. 2010).
Since the initial studies versus placebo that dem-
onstrated the activity of the EGFR TKIs, a number of
2.2 EGFR Tyrosine Kinase Inhibitors studies have been performed that have expanded and
further refined the role of these drugs. Most impor-
The EGFR TKIs were the first targeted therapies to tantly, and as a direct result of the observation that
have become established for the treatment of lung some patients (i.e. scant or never smokers, women,
cancer. Gefitinib, erlotinib, and related quinazoline Asians), experienced truly dramatic benefit in terms
molecules are small molecule inhibitors of the intra- of both radiographic response and progression-free
cellular domain of the EGFR (Woodburn 1999). Only survival, activating mutations (primarily in exons 19
one agent, erlotinib, is fully approved for the treat- and 21) in the EGFR were discovered. Retrospective
ment of NSCLC in the United States. Gefitinib, ini- analyses as well as prospective studies have now been
tially granted approval in the US, is currently performed that confirm the importance of these
approved in many European and Asian countries. The mutations. The Iressa Pan-Asia Study (IPASS)study
initial trials with both agents were negative. The tested gefitinib versus chemotherapy as initial therapy
drugs were combined with standard platinum based in scant or never smokers with advanced NSCLC
chemotherapy in unselected populations as first-line (Mok et al. 2009). There was a clear progression-free
chemotherapy for advanced NSCLC. Four random- survival advantage for gefitinib, though no overall
ized trials with this design were negative (Giaccone survival advantage, apparently due to cross-over.
et al. 2004; Herbst et al. 2004; Gatzemeier et al. 2007; Retrospective analysis demonstrated that the benefit
Herbst et al. 2005). It is an interesting distinction, that was mostly due to treatment of patients with acti-
despite the common target of the two classes of anti- vating mutations. Prospective studies have now been
EGFR agents is the demonstrated benefit of EGFR accomplished in patients with activating mutations
antibodies in addition to cytotoxic chemotherapy in demonstrating unprecedented response rates and
778 M. J. Edelman and N. Ijaz

progression-free survival (Maemondo et al. 2010). 3.1 VEGF Antibodies


At this time, the optimal use of these agents in
advanced disease, i.e. initial use, in maintenance or at Bevacizumab was the first antiangiogenesis agent to
time of relapse in mutated patients is controversial, gain approval by the US FDA and is currently
though the current trend is to employ them as soon as approved for use in five malignancies: breast cancer,
possible. colon cancer, ovarian cancer, glioblastoma multi-
Both EGFR inhibitors are well tolerated compared forme, and NSCLC. It a recombinant humanized
with other antineoplastic agents. Toxicities, including monoclonal antibody that binds and neutralizes
rash and diarrhea, were described for both agents, VEGF, thereby preventing its interaction with the cell
with a somewhat greater frequency described for surface VEGF receptor and blocking the VEGF
erlotinib. However, the rate of treatment discontinu- pathway.
ation for toxicity was very low for both drugs. In the A preliminary randomized phase II study demon-
National Cancer Institute of Canada BR.21 trial, only strated improved survival for patients with advanced
5% of patients discontinued erlotinib treatment for NSCLC treated with bevacizumab15 mg/kg in com-
toxicity compared with 2% of patients in the placebo bination with carboplatin/paclitaxel as initial therapy
arm. Of note, the rate of interstitial lung disease (ILD) (Johnson et al. 2004). Of concern, there were six cases
was very low and possibly unrelated to the use of of significant hemoptysis, four of them fatal. These
erlotinib. Both the patients treated with erlotinib and cases were confined to patients with squamous his-
those treated with placebo experienced a 0.8% inci- tology. This study informed the design of the eastern
dence of ILD. This is in contrast to the Japanese cooperative oncology group (ECOG) 4599 study
experience where uncontrolled trials indicated a 4 to which excluded patients with squamous histology,
10% incidence of ILD, and may represent toxicity brain metastases, anticoagulation or other history of
unique to the Japanese patient population (Lynch bleeding or thrombotic events (Sandler et al. 2006).
et al. 2010; Khambata-Ford et al. 2010). In this selected population, the trial demonstrated
The role of EGFR TKIs in combination with prolonged overall and progression-free survival
radiotherapy is less clear. Preclinical evidence supports bevacizumab was added to carboplatin/paclitaxel.
the potential use of these agents in combination with However, an unplanned subset analysis by the ECOG
radiation (Chinnaiyan et al. 2005). To date, small investigators indicated that patients over 70 years of
studies have demonstrated the feasibility of this age who received bevacizumab experienced increased
approach (Choong et al. 2008; Wang et al. 2011). In myelotoxicity and no survival benefit (Ramalingam
addition to the concomitant use of the agents, another et al. 2008).
approach is to use EGFR TKIs as maintenance treat- The phase III AVAstin In Lung cancer (AVAiL)
ment. SWOG 0023 evaluated gefitinib in patients with trial showed that combining bevacizumab (7.5 or
locally advanced NSCLC with stable or responding 15 mg/kg) with platinum based chemotherapy
disease after concurrent chemoradiation. Surprisingly, improved progression-free survival for ‘‘bevacizumab-
they not only failed to demonstrate an advantage for eligible’’ patients with advanced NSCLC, but not
this approach, but actually demonstrated a significant overall survival (Reck et al. 2009). This study enrolled
decrease in overall survival (Kelly et al. 2008). 1,043 patients who were randomly assigned to receive
cisplatin 80 mg/m2 and gemcitabine 1,250 mg/m2
for up to six cycles plus low-dose bevacizumab
3 Vascular Endothelial Growth Factor (7.5 mg/kg), high-dose bevacizumab (15 mg/kg), or
placebo every 3 weeks until disease progression
VEGF is a potent endothelial angiogenic factor (placebo, n = 347; low-dose, n = 345; high-dose,
expressed in a wide variety of tumors. Aberrant sig- n = 351). For unspecified reasons, the primary end
naling via this pathway is frequently associated with point was amended from overall survival to progres-
neoangiogenesis in tumors. In NSCLC, high levels of sion-free survival while the trial was in progress. PFS
VEGF expression indicate a poor prognosis, sug- was significantly prolonged; the hazard ratios for pro-
gesting that treatment geared toward this pathway gression-free survival were 0.75 (6.7 vs. 6.1 months for
might be effective target for therapy. placebo; P = 0.003) in the low-dose group and 0.82
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 779

(6.5 vs. 6.1 months for placebo; P = 0.03) in the high- chemoradiotherapy. Patients were stratified based
dose group compared with placebo. Objective response upon the risk of toxicity due to bevacizumab. The
rates were 20.1, 34.1, and 30.4% for placebo, low-dose ‘‘high risk’’ stratum (any of the following: squamous
bevacizumab, and high-dose bevacizumab, respec- histology, tumor cavitation, location of tumor near a
tively. However, no significant differences were major vessel or a history of hemoptysis) was closed
observed in overall survival. Interestingly, this trend due to unacceptable toxicity. The University of North
has been noted with bevacizumab in combination with Carolina is performing a Phase I/II Trial of induction
chemotherapy in a number of other settings, including carboplatin/paclitaxel with bevacizumab followed by
in the adjuvant treatment of colorectal cancer and in concurrent thoracic conformal radiation therapy with
breast cancer. The increase in progression-free survival carboplatin/paclitaxel, bevacizumab and erlotinib
without an overall survival advantage may imply that locally advanced NSCLC (NCT00280150). Of the
treatment with bevacizumab results in accelerated first 21 patients enrolled, one experienced a grade 5
repopulation of tumor cells or induction of resistance. hemorrhage. At this time, the trial is still active.
It should also be noted that the mechanism of bev- Though investigations continue, it is clear that the
acizumab is unclear. Though an effect on angiogenesis combination of bevacizumab with chemoradiotherapy
was the basis of its development, some have postulated has the potential for significant toxicity and is unli-
that benefit may result from a normalization of vascu- kely to be of benefit in the broad patient population.
lature and increased delivery of chemotherapy (Tong
et al. 2004). In addition, preclinical evidence indicates
that the benefits of bevacizumab may be dependent 3.2 VEGFR TK Inhibitors
upon the specific chemotherapy regimen employed
(Shaked et al. 2008). Numerous small molecule TKIs have been developed
Overall, in combination with chemotherapy, that inhibit one or more of the VEGF receptors. These
bevacizumab is generally well tolerated. The most include sunitinib, sorafenib, vandetanib, cediranib, and
common adverse events include hypertension and axitinib. Sorafenib and sunitinib have been licensed for
proteinuria. Hemorrhage, though uncommon, can be the treatment of renal cell carcinoma and vandetanib
fatal. Other less common events include reversible has received approval for thyroid cancer. Some, for
posterior leukoencephalopathy syndrome (RPLS) and example sunitinib and axitinib have been demonstrated
organ perforation. to have single agent activity in NSCLC (Socinski et al.
The excitement generated by the positive ECOG 2008; Schiller et al. 2009). However, no phase III trial
4599 study led to several attempts to combine bev- has demonstrated a survival advantage for the concur-
acizumab with radiation and chemoradiotherapy in rent administration of these agents with chemotherapy.
locally advanced lung cancer. The Sarah Cannon In fact, due to increased toxicity, some have been
group evaluated bevacizumab in combination with inferior to placebo. For example, a study comparing
chemotherapy in locally advanced NSCLC and lim- carboplatin/paclitaxel to carboplatin/paclitaxel/so-
ited stage SCLC. The trials were halted after five rafenib demonstrated a median OS for standard therapy
patients experienced tracheoesophageal fistulas or of 10.7 versus 10.6 months for the experimental arm
other life threatening GI toxicity (Spigel et al. 2010). (HR = 1.15; 95% CI, 0.94 to 1.41; P = 0.915). A pre-
In the SCLC trial, 2 of 29 patients developed TE specified exploratory analysis revealed that patients
fistulae (one fatal) and a third died of GI hemorrhage. with squamous cell histology had greater mortality
In the NSCLC trial, 2 of 5 patients developed TE with sorafenib (HR = 1.85; 95% CI, 1.22 to 2.81)
fistulae. The SouthWest Oncology Group (SWOG (Scagliotti et al. 2010). A sequential approach, with
0533) performed a pilot trial combining cisplatin/ initial administration of chemotherapy followed by
etoposide and radiotherapy followed by consolidation single agent sunitinib is currently under investigation
docetaxel and the addition of bevacizumab in locally by the CALGB (CALGB 30607, NCT00693992)
advanced NSCLC (NCT00334815). Bevacizumab The side effects of these agents are similar to bev-
was added in three ways, at the initiation of acizumab and include hypertension, proteinuria,
chemoradiotherapy, at day 15 of chemoradiotherapy increased risk of neutropenia, and RPLS. Some carry
and with docetaxel alone after completion of additional toxicities, such as stomatitits (sunitinib).
780 M. J. Edelman and N. Ijaz

A study of sorafenib in combination with 45 Gy of PARP inhibition occur to a similar degree in both
radiotherapy (3 Gy fractions) in patients with ‘‘poor oxygenated and hypoxic cells.
prognosis NSCLC for whom thoracic radiotherapy is Phase I studies in combination with DNA damag-
indicated’’ has been completed at MD Anderson, but ing chemotherapy agents such as platinums and
the results have not been reported (NCT00543335). temozolamide have been reported and demonstrated
Similarly, a phase I study of sunitinib in combination good tolerability (Plummer et al. 2008). Proof of
with radiotherapy is underway at Thomas Jefferson principle for this approach has come through a ran-
University (NCT00437372). domized phase II trial in previously treated ‘‘triple
negative’’ breast cancer, an entity with frequent
BRCA1/2 deficiency (O’Shaughnessy et al. 2011).
4 PARP Inhibitors A phase III study with a similar regimen
(NCT00938652), but in a different patient population
Perhaps the most exciting agents under development is reportedly negative. However, subset analysis
from the standpoint of their potential for future use appears to confirm the earlier findings and there
with radiation are the poly ADP ribose polymerase seems little doubt that this agent will eventually enter
(PARP ) inhibitors. PARP is an important component routine use. Surprisingly, there are relatively few
of the DNA repair pathway, providing the scaffold for studies evaluating PARP inhibitors in combination
replacing damaged or missing bases. The PARPs, the with radiotherapy. The University of Chicago Con-
most abundant of which is PARP1, are a family of sortium has activated a phase I study combining
about 18 nuclear enzymes that polymerize poly ABT-888 with low-dose weekly carboplatin/paclit-
(adenosine diphosphate–ribose) on substrate proteins axel and radiotherapy regimen for the treatment of
to regulate processes such as DNA repair, gene tran- locally advanced NSCLC. In addition, a phase I study
scription, and chromatin architecture. PARP1 is of ABT-88 in combination with whole brain radiation
involved in the repair of single-strand breaks. Inhi- for solid tumor brain metastases, including lung
bition of PARP results in double-strand breaks, which cancer, is currently in progress (NCT00649207).
normal cells can repair through a process of homol-
ogous recombination. In cells with defective homol-
ogous recombination, such as those with BRCA1or 5 Proteasome Inhibitors
BRCA2 deficiency, PARP inhibition leads to persis-
tent double-strand breaks, inducing cell death through Bortezomib (Velcade, PS-341) represents the first
apoptosis as well as autophagy (Carey and Sharpless proteasome inhibitor to have shown anti-tumor
2011). Preclinical work has demonstrated the poten- activity. The drug is a tripeptide with pyrazinoic acid,
tiation of DNA damaging chemotherapy and radio- phenylalanine and l with boronic acid. The boron
therapy with PARP inhibitors in cell lines (including atom reversibly binds with high-affinity and speci-
the NSCLC line A549) and in a head and neck cancer ficity at the catalytic site of the 26S proteasome that
xenograft model (Calabrese et al. 2004; Khan et al. degrades ubiquitinated proteins. It blocks the nuclear
2010). Utilizing a murine lung cancer xenograft factor-kappa B (NF-kB), leading to increased apop-
model, potentiation of radiotherapy by the PARP tosis, and inhibition of tumor cell adhesion to the
inhibitor ABT-888 was demonstrated with cytotox- stroma along with having anti angiogenic properties.
icity due to both apoptosis and autophagy (Albert While multiple signaling pathways are involved,
et al. 2007). In glioma cell lines the PARP inhibitor proteasome inhibition prevents degradation of pro-
KU-0059436 increased radiosensitivity in a replica- apoptotic factors, leading to programmed cell death in
tion-dependent manner which was enhanced by neoplastic cells.
fractionation (Dungey et al. 2008). The authors Currently, bortezomib is approved for the treat-
hypothesized that PARP inhibition increased the ment of multiple myeloma. Its role in other malig-
incidence of collapsed replication forks after ionizing nancies in general and lung cancer in particular is
radiation, generating persistent DNA double-strand yet to be fully established. Bortezomib has been
breaks and consequent apoptosis. Liu et al. (2008) studied either as a single agent or in combination with
demonstrated that the radiation potentiating effects of various chemotherapy drugs (erlotinib, carboplatin/
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 781

gemcitabine etc.) in lung cancer but has failed to cell lines. There are 18 HDACs generally divided into
demonstrate sufficient activity to warrant further four classes based on homology to yeast HDACs.
studies (Lynch et al. 2009; Davies et al. 2009). HDACs remove the acetyl groups from the lysine
There is considerable preclinical rationale for the residues of histone. They have many protein sub-
combination of bortezomib and other proteasome strates in addition to histones that are involved in
inhibitors with radiation (Edelman 2005). A phase I regulation of gene expression, cell proliferation, and
trial of bortezomib given at days 1, 4, 15, and mitotic cell death. Inhibition of HDACs cause accu-
18 in addition to carboplatin (AUC = 2)/paclitaxel mulation of acetylated forms of these proteins, thus
50 mg/m2 with concurrent radiotherapy (61 Gy over altering their function. Vorinostat (suberoylanilide
6 weeks) as induction treatment for locally advanced hydroxamic acid), is the first HDAC inhibitor
(Stage IIIa, selected IIIb) has been reported as a tri- approved for clinical use in the treatment of the
modality approach to lung cancer (Edelman et al. cutaneous T-cell lymphoma. It inhibits the zinc-con-
2010). Bortezomib was administered during the taining classes I, II, and IV, but does not affect the
6-week induction chemoradiotherapy. Surgical NAD (+)-dependent class III, enzymes.
resection was attempted in the patients with medias- Histone deacetylase inhibitors constitute a prom-
tinal sterilization. Bortezomib was well tolerated, ising treatment for cancer therapy due to their
with no unexpected toxicities during the induction potential wide spectrum of activity and relatively low
phase. However, there were three postoperative single agent toxicity. Randomized phase II and phase
deaths, two from pneumonitis and one from failure of III studies used carboplatin and paclitaxel in combi-
the bronchopulmonary flap and so the trial was halted nation with either vorinostat or placebo for first-line
as a consequence of these toxicities. Although the therapy of advanced NSCLC have been completed. In
relationship of these events to the use of bortezomib the US, an NCI sponsored randomized phase II trial,
was not completely clear, it was felt that the contin- patients with previously untreated stage IIIB or IV
uation of this trial was inappropriate. However, there NSCLC were randomly assigned to carboplatin (AUC
was a high incidence of complete pathologic response 6 mg/ml x min) and paclitaxel (200 mg/m2 day 3)
and so it appears that cautious exploration of this agent with either vorinostat (400 mg by mouth daily) or
in the non-operative setting is appropriate. A non- placebo. Vorinostat or placebo was given on days 1
operative study utilizing a similar regimen through 14 of each 3-week cycle to a maximum of six
(NCT00093756) has been conducted by the North cycles. The response rate was 34% with vorinostat
Central Cancer Treatment Group, but has not yet been versus 12.5% with placebo (P = 0.02). There was a
reported. trend toward improvement in median progression-free
A number of second generation proteasome survival (6.0 vs. 4.1 months; P = 0.48) and overall
inhibitors (e.g., carfilzomib) are currently in devel- survival (13.0 vs. 9.7 months; P = 0.17) in the vori-
opment. Some of these agents are irreversible inhib- nostat arm (Ramalingam et al. 2010a). While the
itors, in contrast to borezomib or have different investigators concluded that this was a potentially
chemical attributes that may convey unique activity promising approach, a company sponsored phase III
(Kuhn et al. 2011; Dick and Fleming 2010). At least trial employing a different schedule was negative.
one (MLN-9708) is oral. No data exist regarding their There are several studies evaluating the role of
potential activity in lung cancer either alone, or with HDAC inhibitors with radiotherapy in NSCLC. A
chemotherapy or radiation. dose-escalation study of single agent vorinostat with
radiation for the palliation of locally advanced
NSCLC is being conducted at Yale (NCT00821951).
6 HDAC Inhibitors A phase I/II study combining vorinostat with carbo-
platin/paclitaxel and XRT in locally advanced disease
Histone deacetylases (HDAC) are a group of novel is in progress at the Fred Hutchison Cancer Research
anticancer agents that have been shown to effect cell Center (NCT00662311). Stanford investigators are
differentiation, proliferation, cell-cycle arrest, and evaluating the role of vorinostat in combination with
apoptosis (Kim and Bae 2011). They also inhibit cell stereotactic radiosurgery for brain metastases due to
migration, invasion, and angiogenesis in many cancer NSCLC (NCT00946673).
782 M. J. Edelman and N. Ijaz

at a number of sites. In addition, everolimus is being


7 mTOR Inhibitors evaluated in combination with whole brain radio-
therapy for the treatment of CNS metastases from
The mammalian target of rapamycin (mTOR) is an NSCLC (NCT00892801).
intracellular serine/threonine protein kinase posi-
tioned at a fundamental point in many cellular sig-
naling pathways and has been identified as a major 8 IGF-IR Inhibitors
culprit in tumorigenesis. Several inhibitors of mTOR,
including temsirolimus, everolimus, and ridaforoli- The insulin-like growth factors (IGFs: IGF-1 and
mus (formerly deforolimus) have been developed and IGF-2) are mediators of growth hormone and they
assessed for their safety and efficacy in patients with play major role in cell growth, differentiation, sur-
cancer. Temsirolimus is an intravenously adminis- vival, and metastasis. The effects of the IGFs are
tered agent approved in the United States for the mediated by the IGF-1 receptor (IGF-1R), a tyrosine
treatment of advanced renal cell carcinoma. Everoli- kinase receptor with homology to the insulin receptor
mus is an oral agent approved for the treatment of (IR). The IGF-binding proteins (IGFBPs) are another
advanced renal cell cancer after failure of treatment component of the IGF system comprising of a class of
with sunitinib or sorafenib. Additionally, there is six soluble secretory proteins. They represent a
recent evidence of significant single agent activity in unique class of naturally occurring IGF-antagonists
pancreatic neuroendocrine tumors (Yao et al. 2011). that bind to and sequester IGF-1 and IGF-2, inhibiting
Ridaforolimus is a newer agent currently under their access to the IGF-1R. Deregulation of the IGF
investigation. Clearly, mTOR inhibitors, either alone system is a well recognized element in the progres-
or in combination with other anticancer agents, have sion of multiple cancers, increasing the tumorigenic
demonstrated activity in cancer and the potential to potential of breast, prostate, lung, colon and head and
provide anticancer activity in numerous tumor types. neck squamous cell carcinoma (Gridelli et al. 2010).
Everolimus demonstrated limited single agent Currently, both antibodies and small molecule
activity in a study of previously treated small cell inhibitors targeting the IGF pathway are in develop-
lung cancer patients (Tarhini et al. 2010). In contrast, ment. Figitumumab (CP-751,871) is a fully human
some promise for everolimus (10 mg daily) was immunoglobin G monoclonal antibody against IGF-
demonstrated in a multicenter phase II trial (n = 85) 1R that has demonstrated promising activity in
of patients with previously treated NSCLC (Soria NSCLC when combined with carboplatin/paclitaxel
et al. 2009). A phase I study evaluating the combination in a phase II trial. It has an effective half-life of
of everolimus and docetaxel for refractory/relapsed approximately 20 days, and it has been generally well
NSCLC has been completed and a phase II trial is tolerated in clinical studies when given alone or in
currently accruing (Ramalingam et al. 2010b). combination with chemotherapy and targeted agents
A trial combining everolimus with gefitinib has been (Karp et al. 2009a). Mild to moderate asymptomatic
completed for patients with relapsed NSCLC hyperglycemia is observed with figitumumab therapy.
(NCT00456833). The 13% partial response rate did not A randomized phase II study compared paclitaxel
appear to be much greater than gefitinib alone and did (200 mg/m2), carboplatin (AUC = 6), and fig-
not warrant further development (Price et al. 2010). itumumab (20 mg/Kg) or paclitaxel and carboplatin
Both agents are under active investigation as a alone for 3 weeks for a total of 6 cycles. The study
potential radiosensitizers in lung cancer. In vitro appeared to demonstrate and advantage for patients
evidence indicates that the PI3kinase/AKT pathway is with squamous cell carcinoma (Karp et al. 2009b).
upregulated with radiotherapy and that inhibition of However a phase III trial in lung cancer was discon-
the mTOR pathway is radiosensitizing (albeit using tinued due to excessive deaths but others are still in
prostate and breast cancer models) (Cao et al. 2006; progress (Jassem et al. 2010). In vitro evidence indi-
Albert et al. 2006). Phase I studies combining cates that inhibition of IGF-1R results in radiosensi-
temsirolimus with radiotherapy (NCT00796796) and tization (Cosaceanu et al. 2004). No studies of these
everolimus with radiotherapy (NCT01167530) che- agents, either alone with radiation or as part of che-
moradiotherapy (NCT01063478) are now in progress moradiotherapy have been reported to date.
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 783

urinary PGE-M (a stable, and major downstream


9 Cyclooxygenase-2 Inhibitors metabolite). These studies also indicate that if COX-2
inhibitors are to have any benefit, that benefit will
Cyclooxygenase-2 (COX-2) is an enzyme of the ara- likely be confined to those with tumors that are driven
chidonic acid cascade. It is upregulated and overex- by COX-2. A phase III study (CALGB 30801) is
pressed in many tumors, including lung cancer as well currently in progress in patients selected for COX-2
as in newly formed blood vessels within tumors. The expression on the basis of immunohistochemistry.
mechanism by which COX-2 overexpression affects Preclinical studies show that COX-2 inhibitors
NSCLC is unclear. It is believed that prostaglandin E2 enhance in vitro cell radiosensitivity and in vivo
(PGE-2), a downstream product of COX-2, promotes tumor radioresponse. MD Anderson investigators
tumor growth and invasion by stimulation of VEGF evaluated the use of celecoxib with radiotherapy in
production, inhibits immune surveillance, and upreg- poor prognosis patients with localized or locally
ulates bcl-2 and various matrix metalloproteinases. advanced NSCLC. The trial had three cohorts: (1)
COX-2 overexpression is considered a marker of poor locally advanced NSCLC with obstructive pneumo-
prognosis in NSCLC (Lee et al. 2007). nia, hemoptysis, and/or minimal metastatic disease
The availability of selective COX-2 inhibitors (e.g., treated with 45 Gy in 15 fractions; (2) inoperable
celecoxib), coupled with the potential role of COX-2 early-stage NSCLC treated with definitive radiation
overexpression, has made COX-2 a rational target in of 66 Gy in 33 fractions; and (3) patients who
NSCLC. Despite considerable preclinical evidence, received induction chemotherapy but who were not
the results of clinical trials in lung cancer have been eligible for concurrent chemoradiotherapy trials.
mixed. Altorki demonstrated that carboplatin/paclit- These patients received 63 Gy in 35 fractions. Cele-
axel when administered pre-operatively could induce coxib was administered orally on a daily basis 5 days
COX-2 and that this effect was abrogated by celecoxib before and throughout the course of radiotherapy with
with an accompanying improvement in response doses escalated from 200, 400, 600, to 800 mg/d. The
(Altorki et al. 2003). However, a large (n = 561) main toxicities were grades 1 and 2 nausea and
phase III study in an advanced disease (NVALT) did esophagitis, and they were independent of the dose of
not demonstrate an advantage for celecoxib when celecoxib or radiotherapy schedule. A maximal
added to carboplatin/docetaxel in advanced disease tolerated dose was not reached in this study. The
(Groen et al. 2009). There was a borderline improve- treatment resulted in local progression-free survival
ment in the objective response rate with celecoxib of 66.0% at 1 year and 42.2% at 2 years. A multi-
compared to placebo (32 vs. 27%). However, there center phase I/II study (RTOG 0213) of celecoxib
was no improvement in either progression-free or plus radiotherapy in locally advanced NSCLC in
overall survival 4.5 versus 4.1 and 8.2 versus patients with intermediate prognostic factors that
8.3 months, respectively. failed to complete accrual. A Dutch randomized
Cancer and Leukemia Group B investigators phase II/III study compared standard radiotherapy for
evaluated inhibitors of two eicosanoid pathways stage II/III patients with celecoxib 400 mg bid (begun
COX-2 (celecoxib) and 5-lipoxygenase (zileuton) 1 week before radiation) versus placebo with the
added to carboplatin/gemcitabine in a randomized drugs continuing for one year. Unfortunately, the
phase II trial (Edelman et al. 2008). Though the study study failed to accrue and was closed when only 41
was negative, a pre-defined analysis found that while patients had been randomized (De Ruysscher et al.
high COX-2 expression was a negative prognostic 2007). There were no significant differences between
marker for overall survival for patients not receiving the arms, however, overall, one year and two year
celecoxib, it was a positive prognostic marker for survival numerically favored the use of celecoxib.
patients with moderate to high levels of COX-2 In a single-institution phase II study concurrent
expression who did receive celecoxib in addition to chemoradiation with paclitaxel and carboplatin
chemotherapy. Multivariate analysis confirmed the alongwith with 400 mg of celecoxib was given-twice-
interaction of COX-2 and celecoxib on survival. This daily until disease progression occurred in patients with
study, and several others utilized either COX-2 stage IIIA or IIIB (Mutter et al. 2009). The overall
expression evaluated by immunohistochemistry or objective response rate seen was 42.9%, and the median
784 M. J. Edelman and N. Ijaz

overall survival time was 203 days. In contrast to EML4-ALK fusion oncogene or its variants have a
nonresponders, those patients with complete and partial phenotype similar to that of EGFR mutation positive
responses had a significant decrease in the level of patients, i.e. adenocarcinoma histology, younger age
urinary PGE-M. Patients with very high levels of PGE- at presentation, and patients with never or light
M before initiation of therapy also responded poorly to smoking history.
therapy. The trial was terminated because it did not A phase I dose-escalation study with crizotinib
meet the predetermined goal of 80% overall response demonstrated substantial activity in patients with
rate. In unselected patients, the addition of celecoxib to advanced NSCLC harboring the EML4/ALK trans-
concurrent chemoradiotherapy with inoperable stage location (Kwak et al. 2010). There was a confirmed
IIIA/B NSCLC did not improve survival. This study radiographic response rate of 57% and a disease
also showed that urinary PGE-M appears to be a control rate of 87% at eight weeks. The estimated
promising biomarker for predicting response to COX-2 probability of progression-free survival at six months
inhibition in NSCLC (Liao et al. 2005). was 72%. The most common side effects were fati-
At this time apricoxib, a new more potent selective gue, nausea/vomiting, diarrhea, and visual distur-
COX-2 inhibitor is currently under investigation in a bances associated with the transition from dark to
variety of malignancies, including NSCLC (Edelman light. There are currently trials evaluating crizotinib
2011). These studies select patients based upon uri- in first-line, second line and subsequent therapy.
nary PGE-M suppression. If there is to be a future for Approval of the agent for routine clinical use is likely
COX-2 inhibitors in lung cancer, either in combina- to occur within the next 1–2 years. As the agent is
tion with chemotherapy or radiation, it is likely that it also an inhibitor of c-met, it may also find a role in
will emerge from the studies currently underway in patients with EGFR mutations who have developed
these enriched populations. resistance to EGFR TKIs as a consequence of c-met
amplification. As c-met inhibitors may potentiate
radiotherapy either through direct toxicity or an
10 ALK Targeted Inhibitors antiangiogenic mechanism, crizotinib (as the first
agent to be approved with this activity) should be
Transforming rearrangements of the anaplastic lym- evaluated in this role (Welsh et al. 2009; Hu et al.
phoma kinase (ALK) gene have recently been 2009).
described in NSCLC. A certain subset of patients with
NSCLC have an inversion in chromosome 2 that
juxtaposes the 50 end of the echinoderm microtubule- 11 Aurora Kinase and Polo-like
associated protein-like 4 (EML4) gene with the 30 end Kinase Inhibitors
of the ALK gene, resulting in the fusion oncogene
EML4-ALK. At least seven ALK gene rearrangement A number of recently described proteins that form a
variants have been described involving different ‘‘mitotic spindle apparatus’’ that synchronizes the
EML4-ALK breakpoints or rarely other non-EML4 movement of tubulin and consequently chromosomal
fusion partners. ALK rearrangements may be identi- segregation. These proteins include the aurora kina-
fied in tumor tissue by reverse transcription-poly- ses, polo-like kinases, kinesin spindle proteins, sur-
merase chain reaction or fluorescent in situ vivin and others. In addition to the function of
hybridization. The orally administered small molecule chromosomal segregation, cytokinesis and other key
tyrosine kinase inhibitor crizotinib (PF02341066), cellular functions mediated by tubulin are also gov-
originally under development as an inhibitor of c-met, erned by these proteins (Andrews et al. 2003).
is also a potent inhibitor of ALK phosphorylation and Overexpression of these proteins, particularly the
signal transduction. This inhibition is associated with aurora kinases has been demonstrated to be associated
G1-S phase cell-cycle arrest and induction of apop- with malignancy in general and lung cancer in par-
tosis in positive cells in vitro and in vivo. ticular (Smith et al. 2005). There are three types of
Crizotinib has demonstrated dramatic antitumor aurora kinase, A, B, and C. Aurora kinase A and B are
benefit with little toxicity and is well tolerated in essential for normal progression of all types of cells
patients with ALK-rearranged NSCLC. Tumors with through mitosis. Aurora C is expressed exclusively in
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 785

the testis and co-localizes with aurora-B. Aurora-A Hh target genes. In the presence of Hh ligands (Shh, Ihh
dysregulation is commonly associated with the or Dhh), PTC is unable to affect SMO signaling, leading
development of cancer. Interestingly, amplification of to enhanced formation of activated forms of Gli (GliA)
aurora-A has been demonstrated to result in resistance and upregulation of Hh target genes. Gene mutations
to paclitaxel as well as radiation (Anand et al. 2003; (PTC, SMO) or abnormal over expression of Hh
Guan et al. 2007). ligands may lead to elevated expression of Hh target
A plethora of inhibitors to the aurora kinases (both genes. Abnormalities of the Hh pathway occur in both
selective for A or B as well as dual inhibitors) and NSCLC and SCLC, but seem to be particularly com-
polo-like kinases have entered clinical trials, Table 1. mon in small cell lung cancer (Gialmanidis et al. 2009;
Single agent toxicity data have been relatively Vestergaard et al. 2006).
similar, with both classes of agents limited by dose Cyclopamine is a naturally occurring steroidal
dependent neutropenia. There is considerable ratio- alkaloid which inhibits Hh ligand dependent and
nale for evaluating these agents in combination with independent pathways via direct interaction with Smo
chemotherapy and radiation. Polo-like kinases are (Kubo et al. 2004). GDC-0449 is an orally administered
involved in DNA damage repair and inhibition has analogue of cyclopamine which has shown activity and
been demonstrated to result in enhancement of radi- excellent tolerability as a single agent in tumors har-
ation and chemotherapy cytotoxicity (Gerster et al. boring mutations associated with activation of the Hh
2010; Zhou and Bai 2004). Aurora-B kinase inhibitors signaling pathway (i.e, basal cell carcinoma, medullo-
have been demonstrated to sensitize cells to radiation blastoma) (Von Hoff et al. 2009; Rudin et al. 2009). The
(Tao et al. 2009). Sensitization to chemotherapy and agent demonstrated a favorable pharmacokinetic pro-
radiotherapy may be schedule dependent and clinical file, with high, sustained, plasma concentrations and a
evaluation of these agents will require careful atten- terminal half-life of greater than seven days. No dose-
tion to the timing of administration relative to radia- limiting adverse events were observed at the three dose
tion and/or chemotherapy. At this time, there is levels (150, 270, or 540) studied.
relatively little data regarding activity in lung cancer Currently, there are several trials underway in lung
and no clinical studies have yet evaluated the agents cancer. GDC-0449 is being tested by the ECOG in
in combination with radiotherapy. combination with cisplatin/etoposide in extensive
small cell lung cancer (NCT00887159). No trials are
currently underway combining these agents with
12 Hedgehog Pathway Inhibitors radiotherapy. However, such studies are logical as the
Hh pathway is upregulated in response to radiation
The Sonic Hedgehog (Shh) pathway plays an impor- and is therefore a logical target (Chen et al. 2011).
tant role in embryogenesis, stem cell maintenance,
tissue repair, and tumorigenesis (Yang et al. 2010).
Intercellular signaling proteins of the Hh family have 13 Radiopharmaceuticals
come to be recognized as mediators of fundamental
processes in embryonic development and is a major A relatively unexplored approach in lung cancer is the
regulator for stem cell maintenance, cell differentia- use of radiopharmaceuticals. Broadly defined, these
tion, tissue polarity, and cell proliferation. The key are agents with a specific targeting moiety coupled to
molecules in this pathway include: (1) smoothened a radionuclide. Table 2 describes the basic terminol-
(SMO)—a seven member transmembrane domain, (2) ogy for radiopharmaceuticals and some of the char-
patched (PTC)—a transmembrane protein which acteristics of the most widely evaluated radionuclides
serves at the receptor for Hh, (3) Gli—a family of (Milenic and Brechbiel 2004). Part of the attraction of
downstream transcription factors that regulate target these agents is that this approach allows for a real
gene expression by direct association with a specific time assessment of the presence of a target through
consensus sequence located in the promoter region of the use of related imaging diagnostics or in some
the target genes. In the absence of Hh, the Hh receptor cases the agent itself. This approach has been vali-
PTC suppresses SMO activity and repressor forms of dated for the treatment of B-cell non-Hodgkins
Gli (GliR) are generated, leading to downregulation of lymphoma where two agents (yttrium 90 ibritumomab
786 M. J. Edelman and N. Ijaz

Table 2 Radionuclide terminology and examples


Term Definition Examples Half-life Tissue range (mm) Energy (mEv)
Alpha particle 2 protons ? 2 neutrons (He nucleus) Bi211 46 min 0.04–0.1 5.87
212
Bi 1h 0.04–0.1 6.09
At211 7.2 h 0.04–0.1 5.87
Beta particle electrons Yt90 2.7days 2.76 2.3
a
Re188 17 h 2.43 2.1
a
Lu177 6.7days 0.4 0.50
a 131
I 8days 0.28 0.81
a
also emits gamma radiation (photons)
Adapted from Millenic and Brechibel (Price et al. 2010)

and iodine-131 tositumomab) have been approved. gamma (photon) as well as beta emitter have the
Both agents couple a radioisotope to a CD-20 anti- advantage of the potential for dual use as both as an
body, which is highly specific for B-cells. imaging as well as a therapeutic agent. A typical
In general, all agents share certain features. There paradigm is to administer either a small dose of the
is a targeting moiety, such as an antibody or a agent (if a gamma emitter) or a Tc99 labeled version
receptor analogue, coupled to a molecule capable of of the agent to determine the avidity of the target
binding the radionuclide chosen. Binding can be as well as the distribution which allows for determi-
directly to the protein (halogenation) at a tyrosine nation of the dose based upon the target uptake as
group, or through chelating agents that bind to the well as the potential limits of normal tissues.
protein and to the radionuclide. Most radionuclides In solid tumor oncology there are relatively few
for therapeutic purposes are either alpha or beta tumor specific surface antigens that lend themselves
emitters. The characteristics of the radionuclide are to this approach. One such specific antigen is the
critical determinants of potential applicability and somatostatin receptor family. There are at least five
activity. Too short a half-life will prohibit generalized SSTRs that can be exploited for this purpose. SSTR2
use of the agent. Too long a half-life increases the is frequently expressed in both small cell and NSCLC
potential toxicity to the patient and others. The path as well as on peritumoral neovasculature (O’Byrne
length and energy imparted by the radionuclide will et al. 2001).
determine the potential activity and toxicity. Beta P2045 is an agent that couples Re188 to a somato-
emitters generally deposit energy at some distance statin fragment specific for SSTR2. The approach had
from the decay event. However, the amount of energy its origins in a Tc99 labeled diagnostic agent developed
is relatively low. In comparison, the alpha emitters prior to the advent of FDG-PET that demonstrated good
deliver a large amount of energy, but the path length ability to distinguish between benign and malignant
is short. Longer path lengths allow for effects on solitary pulmonary nodules on the basis of the presence
tumor cells that may not express the target, too long a of somatostatin receptors. A phase I trial has been
path length increases the risk of bystander organ reported that demonstrated the feasibility and tolera-
toxicity. Most agents that have entered clinical eval- bility of this approach (Edelman et al. 2009). However,
uation are renally excreted and hence the kidney the study was terminated due to concerns regarding the
becomes the dose-limiting organ, though some degree potential long-term renal toxicity of the agent. Given
of marrow toxicity is also frequently observed. the poor prognosis for advanced lung cancer patients,
There are several unique aspects to the use of ra- this was probably excessively cautious and develop-
diopharmaceuticals including the ability to image the ment of this agent is to be resumed in the near future.
tumor (which can assist with determining the appro- A number of similar molecules have also been
priateness of treatment) as well as determining the developed. The somatostatin receptor is frequently
dose. For the two licensed agents, the radionuclide is expressed on neuroendocrine carcinomas. Conse-
a beta emitter. Agents in which the radionuclide is a quently, small cell lung cancer has been a disease of
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 787

particular interest for this approach. Studies in a Calabrese CR, Almassy R, Barton S et al (2004) Anticancer
murine small cell lung cancer model have been chemosensitization and radiosensitization by the novel
poly(ADP-ribose) polymerase-1 inhibitor AG14361. J Natl
reported that compared the unlabeled somatostatin Cancer Inst 96(1):56–67
analogue with the radionuclide labeled agent dem- Cao C, Subhawong T, Albert JM, Kim KW, Geng L, Sekhar
onstrating considerable activity utilizing the radiola- KR, Gi YJ, Lu B (2006) Inhibition of mammalian target of
beled agent in contrast to no activity for the parent rapamycin or apoptotic pathway induces autophagy and
radiosensitizes PTEN null prostate cancer cells. Cancer Res
compound (Schmitt et al. 2004). 66(20):10040–10047
Other targeting molecules can clearly be utilized, Cappuzzo F, Ciuleanu T, Stelmakh L (2010) Erlotinib as
though relatively few have been developed. Of par- maintenance treatment in advanced non-small-cell lung
ticular relevance to lung cancer, an EGFR antibody cancer: a multicentre, randomised, placebo-controlled phase
3 study. Lancet Oncol 11(6):521–529
labeled with Re188 has been developed (Chopra Carey LA, Sharpless NE (2011) PARP and cancer—if it’s
2008). Clinical experience outside of the somatostatin broke, don’t fix it. N Engl J Med 364:277–279
analogues however, is quite limited. In addition, most Chen YJ, Lin CP, Hsu ML, Shieh HR, Chao NK, Chao KS
agents that have entered the clinic have been tested (2011) Sonic hedgehog signaling protects human hepato-
cellular carcinoma cells against ionizing radiation in an
utilizing a single administration. Relatively few have autocrine manner. Int J Radiat Oncol Biol Phys 80(3):
been evaluated for repeated dosing, the most common 851–859 [Epub ahead of print]
approach for both standard radiotherapy as well as Chinnaiyan P, Huang S, Vallabhaneni G, Armstrong E,
chemotherapy. In addition, combination with che- Varambally S, Tomlins SA, Chinnaiyan AM, Harari PM
(2005) Mechanisms of enhanced radiation response follow-
motherapy agents has only rarely been explored, even ing epidermal growth factor receptor signaling inhibition by
in preclinical models. erlotinib (Tarceva). Cancer Res 65(8):3328–3335
Choong NW, Mauer AM, Haraf DJ et al (2008) Phase I trial of
erlotinib-based multimodality therapy for inoperable stage III
non-small cell lung cancer. J Thorac Oncol 3(9):1003–1011
References Chopra A (2008) 188Re-labeled humanized monoclonal anti-
epidermal growth factor receptor antibody.Molecular ima-
Albert JM, Kim KW, Cao C, Lu B (2006) Targeting the Akt/ ging and contrast agent database (MICAD) [Internet].
mammalian target of rapamycin pathway for radiosensiti- Bethesda (MD): National Center for Biotechnology Infor-
zation of breast cancer. Mol Cancer Ther 5(5):1183–1189 mation (US), 2004–2010. 2008 August 04 [updated 2008
Albert JM, Cao C, Kim KW et al (2007) Inhibition of September 02]
poly(ADP-ribose) polymerase enhances cell death and Cosaceanu D, Iliescu G, Castro J et al (2004) In vitro
improves tumor growth delay in irradiated lung cancer enhancement of response to radiation of human lung cancer
models. Clin Cancer Res 13(10):3033–3042 cells following insulin-like growth factor 1 receptor inac-
Altorki NK, Keresztes RS, Port JL et al (2003) Celecoxib, a tivation. Proc Amer Assoc Cancer Res 45:1 Abstract #1299
selective cyclo-oxygenase-2 inhibitor, enhances the Davies AM, Chansky K, Lara PN Jr, Gumerlock PH, Crowley J,
response to preoperative paclitaxel and carboplatin in Albain KS, Vogel SJ, Gandara DR, Southwest Oncology
early-stage non-small-cell lung cancer. J Clin Oncol Group (2009) Bortezomib plus gemcitabine/carboplatin as
21(14):2645–2650 first-line treatment of advanced non-small cell lung cancer:
Anand S, Penrhyn-Lowe S, Venkitaraman AR (2003) Aurora-A a phase II southwest oncology group study (S0339). J Thorac
amplification overrides the mitotic spindle assembly check- Oncol 4:87–92
point, inducing resistance to Taxol. Cancer Cell 3:51–62 De Ruysscher D, Bussink J, Rodrigus P, Kessels A, Dirx M,
Andrews PD, Knatko E, Moore WJ, Swedlow JR (2003) Houben R, Wanders R (2007) Concurrent celecoxib versus
Mitotic mechanics: the auroras come into view. Curr Opin placebo in patients with stage II-III non-small cell lung
Cell Biol 15:672–683 cancer: a randomised phase II trial. Radiother Oncol
Baselga J, Norton L, Masui H, Pandiella A, Coplan K, Miller 84(1):23–25
WH Jr, Mendelsohn J (1993) Antitumor effects of doxoru- Dick LR, Fleming PE (2010) Building on bortezomib: second-
bicin in combination with anti-epidermal growth factor generation proteasome inhibitors as anti-cancer therapy.
receptor monoclonal antibodies. J Nat Cancer Inst 85: Drug Discov Today 15(5–6):243–249 Epub 2010 January 29
1327–1333 Dungey FA, Löser DA, Chalmers AJ (2008) Replication-
Blumenschein GR Jr, Paulus R, Curran WJ et al (2011) Phase II dependent radiosensitization of human glioma cells by
study of cetuximab in combination with chemoradiation in inhibition of poly(ADP-Ribose) polymerase: mechanisms
patients with stage III A/B non-small-cell lung cancer: and therapeutic potential. Int J Radiat Oncol Biol Phys
RTOG 0324. J Clin Oncol 29(17):2312–2318 72(4):1188–1197
Bonner JA, Harari PM, Giralt J et al (2006) Radiotherapy plus Edelman MJ (2005) The potential role of bortezomib in
cetuximab for squamous-cell carcinoma of the head and chemoradiotherapy of non-small cell lung cancer. Clin Lung
neck. NEJM 354(6):567–578 Cancer 7(2):S64–S66
788 M. J. Edelman and N. Ijaz

Edelman MJ (2011) Apricoxib: a novel COX-2 inhibitor for Hu SY, Duan HF, Li QF, Yang YF, Chen JL, Wang LS, Wang H
oncology. Drugs of the future (in press) (2009) Hepatocyte growth factor protects endothelial cells
Edelman MJ, Watson D, Xiaofei W et al (2008) Eicosanoid against gamma ray irradiation-induced damage. Acta Phar-
modulation in advanced lung cancer: cyclooxygenase-2 macol Sin 30(10):1415–1420
expression is a positive predictive factor for cele- Jassem J, Langer CJ, Karp DD et al (2010) Randomized, open
coxib ? chemotherapy–cancer and Leukemia Group B trial label, phase III trial of figitumumab in combination with
30203. J Clin Oncol 26:848–855 paclitaxel and carboplatin versus paclitaxel and carboplatin
Edelman MJ, Clamon G, Kahn D, Magram M, Line BR (2009) in patients with non-small cell lung cancer. J Clin Oncol
Targeted radiopharmaceutical therapy for advanced lung 28(15S):539a (abstract 7500)
cancer: phase I trial of rhenium Re188 somatostatin Johnson DH, Fehrenbacher L, Novotny WF (2004) Random-
analogue P2045. J Thor Oncol 4(12):1550–1554 ized phase II trial comparing bevacizumab plus carboplatin
Edelman MJ, Burrows W, Krasna MJ, Suntharalingam M and paclitaxel with carboplatin and paclitaxel alone in
(2010) Phase I trial of carboplatin/paclitaxel/bortezomib previously untreated locally advanced or metastatic non-
and concurrent radiotherapy followed by surgical resection small-cell lung cancer. J Clin Oncol 22(11):2184–2191
in stage III non-small cell lung cancer. Lung Cancer 68(1): Karp DD, Pollak MN, Cohen RB, Eisenberg PD et al (2009a)
84–88 Safety, pharmacokinetics, and pharmacodynamics of the
Gatzemeier U, Pluzanska A, Szczesna A et al (2007) Phase III insulin-like growth factor type 1 receptor inhibitor fig-
study of erlotinib in combination with cisplatin and itumumab (CP-751, 871) in combination with paclitaxel and
gemcitabine in advanced non-small-cell lung cancer: the carboplatin. J Thorac Oncol 4:1397–1403
tarceva lung cancer investigation trial. J Clin Oncol 25: Karp DD, Paz-Ares LG, Novello S et al (2009b) Phase II study
1545–1552 of the anti-insulin-like growth factor type 1 receptor
Gerster K, Shi W, Ng B, Yue S, Ito E, Waldron J, Gilbert R, antibody CP-751, 871 in combination with paclitaxel and
Liu FF (2010) Targeting polo-like kinase 1 enhances carboplatin in previously untreated, locally advanced, or
radiation efficacy for head-and-neck squamous cell carci- metastatic non-small-cell lung cancer. J Clin Oncol 27(15):
noma. Int J Radiat Oncol Biol Phys 77:253–260 2516–2522
Giaccone G, Herbst RS, Manegold C et al (2004) Gefitinib in Kelly K, Chansky K, Gaspar LE et al (2008) Phase III trial of
combination with gemcitabine and cisplatin in advanced maintenance gefitinib or placebo after concurrent chemora-
non-small-cell lung cancer: a phase III trial—INTACT 1. diotherapy and docetaxel consolidation in inoperable stage
J Clin Oncol 22:777–784 III non-small-cell lung cancer: SWOG S0023. J Clin Oncol
Gialmanidis IP, Bravou V, Amanetopoulou SG, Varakis J, 26(15):2450–2456 Epub 2008 March 31
Kourea H, Papadaki H (2009) Overexpression of hedgehog Khambata-Ford S, Harbison CT, Hart LL et al (2010) Analysis
pathway molecules and FOXM1 in non-small cell lung of potential predictive markers of cetuximab benefit in
carcinomas. Lung Cancer 66:64–74 BMS099, a phase III study of cetuximab and first-line
Govindan R, Bogart J, Wang X et al (2009) Phase II study of taxane/carboplatin in advanced non-small-cell lung cancer.
pemetrexed, carboplatin and thoracic radiation with or J Clin Oncol 28(6):918–927
without cetuximab in patients with locally advanced Khan K, Araki K, Wang D et al (2010) Head and neck cancer
unresectable non-small cell lung cancer. Proc Am Soc Clin radiosensitization by the novel poly(ADP-ribose) polymer-
Oncol 27:383s (abstract 7505) ase inhibitor GPI-15427. Head Neck 32(3):381–391
Gridelli C, Rossi A, Bareschino MA, Schettino C, Sacco PC, Kim HJ, Bae SC (2011) Histone deacetylase inhibitors:
Maione P (2010) The potential role of insulin-like growth factor molecular mechanisms of action and clinical trials as anti-
receptor inhibitors in the treatment of advanced non-small cell cancer drugs. Am J Transl Res 3(2):166–179 Epub 2010
lung cancer. Expert Opin Investig Drugs 19(5):631–639 December 26
Groen H, Hochstenbag MM, van Putten JW et al (2009) A Kubo M, Nakamura M, Tasaki A et al (2004) Hedgehog
randomized placebo-controlled phase III study of docetaxel/ signaling pathway is a new therapeutic target for patients
carboplatin with celecoxib in patients (pts) with advanced with breast cancer. Cancer Res 64:6071–6074
non-small cell lung cancer (NSCLC): the NVALT-4 study. Kuhn DJ, Orlowski RZ, Bjorklund CC (2011) Second
J Clin Oncol 27:15 (suppl; abstr 8005) generation proteasome inhibitors: carfilzomib and immuno-
Guan Z, Wang XR, Zhu XF, Huang XF, Xu J, Wang LH, proteasome-specific inhibitors (IPSIs). Curr Cancer Drug
Wan XB, Long ZJ, Liu JN, Feng GK, Huang W, Zeng YX, Targets 11(3):285–295
Chen FJ, Liu Q (2007) Aurora-A, a negative prognostic Kwak EL, Bang YJ, Camidge DR et al (2010) Anaplastic
marker, increases migration and decreases radiosensitivity lymphoma kinase inhibition in non-small-cell lung cancer.
in cancer cells. Cancer Res 67(21):10436–10444 N Engl J Med 363(18):1693–1703 Erratum in: N Engl J
Herbst RS, Giaccone G, Schiller JH et al (2004) Gefitinib in Med 2011 February 10;364(6):588
combination with paclitaxel and carboplatin in advanced Lee JM, Mao JT, Krysan K, Dubinett SM (2007) Significance
non-small-cell lung cancer: a phase III trial—INTACT 2. of cyclooxygenase-2 in prognosis, targeted therapy and
J Clin Oncol 22:785–794 chemoprevention of NSCLC. Future Oncol 3(2):149–153
Herbst RS, Prager D, Fehrenbacher L et al (2005) TRIBUTE: a Liao Z, Komaki R, Milas L, Yuan C, Kies M, Chang JY,
phase III trial of erlotinib hydrochloride (OSI-774) com- Jeter M, Guerrero T, Blumenschien G, Smith CM, Fossella
bined with carboplatin and paclitaxel chemotherapy in F, Brown B, Cox JD (2005) A phase I clinical trial
advanced non-small-cell lung cancer. J Clin Oncol 23: of thoracic radiotherapy and concurrent celecoxib for
5892–5899 patients with unfavorable performance status inoperable/
Novel Targeted Agents and Radiopharmaceuticals in Lung Cancer 789

unresectable non–small cell lung cancer. Clin Cancer Res Price KA, Azzoli CG, Krug LM, Pietanza MC, Rizvi NA, Pao
11(9):3342–3348 W, Kris MG, Riely GJ, Heelan RT, Arcila ME, Miller VA
Liu SK, Coackley C, Krause M, Jalali F, Chan N, Bristow RG (2010) Phase II trial of gefitinib and everolimus in advanced
(2008) A novel poly(ADP-ribose) polymerase inhibitor, non-small cell lung cancer. J Thorac Oncol 5(10):
ABT-888, radiosensitizes malignant human cell lines under 1623–1629
hypoxia. Radiother Oncol 88(2):258–268 Ramalingam SS, Dahlberg SE, Langer CJ (2008) Outcomes for
Lynch TJ, Bell DW, Sordella R et al (2004) Activating elderly, advanced-stage non small-cell lung cancer patients
mutations in the epidermal growth factor receptor underly- treated with bevacizumab in combination with carboplatin
ing responsiveness of non-small-cell lung cancer to gefiti- and paclitaxel: analysis of eastern cooperative oncology
nib. N Engl J Med 350:2129–2139 group rial 4599. J Clin Oncol 26(1):60–65
Lynch TJ, Fenton D, Hirsh V, Bodkin D, Middleman EL, Ramalingam SS, Maitland ML, Frankel P et al (2010a)
Chiappori A, Halmos B, Favis R, Liu H, Trepicchio WL, Carboplatin and paclitaxel in combination with either
Eton O, Shepherd FA (2009) A randomized phase 2 study of vorinostat or placebo for first-line therapy of advanced
erlotinib alone and in combination with bortezomib in non-small-cell lung cancer. J Clin Oncol 28:56–62
previously treated advanced non-small cell lung cancer. Ramalingam SS, Harvey RD, Saba N, Owonikoko TK, Kauh J,
J Thorac Oncol 4:1002–1009 Shin DM, Sun SY, Strychor S, Tighiouart M, Egorin MJ, Fu H,
Lynch TJ, Patel T, Dreisbach L (2010) Cetuximab and first-line Khuri FR (2010b) Phase 1 and pharmacokinetic study of
taxane/carboplatin chemotherapy in advanced non-small- everolimus, a mammalian target of rapamycin inhibitor, in
cell lung cancer: results of the randomized multicenter combination with docetaxel for recurrent/refractory nonsmall
phase III trial BMS099. J Clin Oncol 28(6):911–917 Epub cell lung cancer. Cancer 116(16):3903–3909
2010 January 25 Reck M, von Pawel J, Zatloukal P et al (2009) Phase III trial of
Maemondo M, Inoue A, Kobayashi K (2010) Gefitinib or cisplatin plus gemcitabine with either placebo or bev-
chemotherapy for non-small-cell lung cancer with mutated acizumab as first-line therapy for nonsquamous non-small-
EGFR. N Engl J Med 362(25):2380–2388 cell lung cancer: AVAil. J Clin Oncol 27(8):1227–1234
Mendelsohn J, Kawamoto T, Sato G, Sato J (1990) Hybrid cell Epub 2009 Feb 2. Erratum in: J Clin Oncol 2009;27:2415
lines that produce monoclonal antibodies to epidermal Rudin CM, Hann CL, Laterra J et al (2009) Treatment of
growth factor receptor. United States Patent #4,943,533. medulloblastoma with hedgehog pathway inhibitor GDC-
July 24 0449. N Engl J Med 361:1173–1178
Milenic DE, Brechbiel MW (2004) Targeting of radio-isotopes Sandler A, Gray R, Perry MC et al (2006) Paclitaxel–
for cancer therapy. Cancer Biol Ther 3(4):361–370 carboplatin alone or with bevacizumab for non-small-cell
Mok TS, Wu YL, Thongprasert S et al (2009) Gefitinib or lung cancer. N Engl J Med 355(24):2542–2550 Erratum in:
carboplatin–paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2007;356:318
N Engl J Med 361(10):947–957 Epub 2009 August 19 Scagliotti G, Novello S, von Pawel J (2010) Phase III study of
Mutter R, Lu B, Carbone DP, Csiki I, Moretti L, Johnson DH, carboplatin and paclitaxel alone or with sorafenib in
Morrow JD, Sandler AB, Shyr Y, Ye F, Choy H (2009) A advanced non-small-cell lung cancer. J Clin Oncol
phase II study of celecoxib in combination with paclitaxel, 28(11):1835–1842 Epub 2010 March 8
carboplatin, and radiotherapy for patients with inoperable Schiller JH, Larson T, Ou SH et al (2009) Efficacy and safety of
stage III A/B non-small cell lung cancer. Clin Cancer Res axitinib in patients with advanced non-small-cell lung
15(6):2158–2165 cancer: results from a phase II study. J Clin Oncol
O’Byrne KJ, Schally AV, Thomas A, Carney DN, Steward WP 27(23):3836–3841 Epub 2009 July 13
(2001) Somatostatin, its receptors and analogs, in lung Schmitt A, Bernhardt P, Nilsson O et al (2004) Radiation
cancer. Chemother 47(Suppl 2):78–108 therapy of small cell lung cancer with 177Lu-DOTA-
O’Shaughnessy J, Osborne C, Pippen JE et al (2011) Iniparib Tyr3-octreotate in an animal model. J Nucl Med 45:
plus chemotherapy in metastatic triple-negative breast 1542–1548
cancer. N Engl J Med 364:205–214 Shaked Y, Henke E, Roodhart JM et al (2008) Rapid
Paez JG, Janne PA, Lee JC et al (2004) EGFR mutations in chemotherapy-induced acute endothelial progenitor cell
lung cancer: correlation with clinical response to gefitinib mobilization: implications for antiangiogenic drugs as
therapy. Sci 304:1497–1500 chemosensitizing agents. Cancer Cell 14(3):263–273
Pirker R, Pereira JR, Szczesna A (2009) Cetuximab plus Shepherd FA, Rodrigues Pereira J, Ciuleanu T et al (2005)
chemotherapy in patients with advanced non-small-cell lung Erlotinib in previously treated non-small-cell lung cancer.
cancer (FLEX): an open-label randomised phase III trial. N Engl J Med 353:123–132
Lancet 373(9674):1525–1531 Smith SL, Bowers NL, Betticher DC et al (2005) Overexpres-
Pirker R, Paz Ares L, Eberhardt W et al (2011) Epidermal sion of aurora-B kinase (AURKB) in primary non-small cell
growth factor receptor expression as a predictor of survival lung carcinoma is frequent, generally driven from one allele
for first-line chemotherapy plus cetuximab in FLEX study and correlates with the level of genetic instability. Br J
patients with advanced non-small cell lung cancer. J Thor Cancer 93:719–729
Oncol 6(Suppl 2):S276 Socinski MA, Novello S, Brahmer JR et al (2008) Multicenter,
Plummer R, Jones C, Middleton M (2008) Phase I study of the phase II trial of sunitinib in previously treated, advanced
poly(ADP-ribose) polymerase inhibitor, AG014699, in non-small-cell lung cancer. J Clin Oncol 26(4):650–656
combination with temozolomide in patients with advanced Soria JC, Shepherd FA, Douillard JY et al (2009) Efficacy of
solid tumors. Clin Cancer Res 14(23):7917–7923 everolimus (RAD001) in patients with advanced NSCLC
790 M. J. Edelman and N. Ijaz

previously treated with chemotherapy alone or with che- Von Hoff DD, Lorusso PM, Rudin CM et al (2009) Inhibition
motherapy and EGFR inhibitors. Ann Oncol 20:1674–1681 of the hedgehog pathway in advanced basal-cell carcinoma.
Spigel DR, Hainsworth JD, Yardley DA, Raefsky E, Patton J, N Engl J Med 361:1164–1172
Peacock N, Farley C, Burris HA 3rd, Greco FA (2010) Wang J, Xia TY, Wang YJ et al (2011) Prospective study of
Tracheoesophageal fistula formation in patients with lung epidermal growth factor receptor tyrosine kinase inhibitors
cancer treated with chemoradiation and bevacizumab. J Clin concurrent with individualized radiotherapy for patients
Oncol 28(1):43–48 Epub 2009 November 9 with locally advanced or metastatic non-small-cell lung
Tao Y, Leteur C, Calderaro J, Girdler F, Zhang P, Frascogna V, cancer. Int J Radiat Oncol Biol Phys [Epub ahead of print]
Varna M, Opolon P, Castedo M, Bourhis J, Kroemer G, Welsh JW, Mahadevan D, Ellsworth R, Cooke L, Bearss D,
Deutsch E (2009) The aurora-B kinase inhibitor AZD1152 Stea B (2009) The c-Met receptor tyrosine kinase inhibitor
sensitizes cancer cells to fractionated irradiation and MP470 radiosensitizes glioblastoma cells. Radiat Oncol
induces mitotic catastrophe. Cell Cycle 8(19):3172–3181 22:69
Epub 2009 October 4 Woodburn JR (1999) The epidermal growth factor receptor and
Tarhini A, Kotsakis A, Gooding W, Shuai Y, Petro D, its inhibition in cancer therapy. Pharmacol Ther 82:241–250
Friedland D, Belani CP, Dacic S, Argiris A (2010) Phase Yang L, Xie G, Fan Q, Xie J (2010) Activation of the
II study of everolimus (RAD001) in previously treated small hedgehog-signaling pathway in human cancer and the
cell lung cancer. Clin Cancer Res 16(23):5900–5907 clinical implications. Oncogene 29(4):469–481 Epub 2009
Tong RT, Boucher Y, Kozin SV, Winkler F, Hicklin DJ, November 23
Jain RK (2004) Vascular normalization by vascular endo- Yao JC, Shah MH, Ito T et al (2011) Everolimus for advanced
thelial growth factor receptor 2 blockade induces a pressure pancreatic neuroendocrine tumors. RAD001 in advanced
gradient across the vasculature and improves drug penetra- neuroendocrine tumors, third trial (RADIANT-3) study
tion in tumors. Cancer Res 64(11):3731 group. N Engl J Med 364(6):514–523
Vestergaard J, Pedersen MW, Pedersen N, Ensinger C, Zhou Q, Bai M, Su Y (2004) Effect of antisense RNA targeting
Tümer Z, Tommerup N, Poulsen HS, Larsen LA (2006) polo-like kinase 1 on cell cycle and proliferation in A549
Hedgehog signaling in small-cell lung cancer: frequent in cells. Chin Med J 117:1642–1649
vivo but a rare event in vitro. Lung Cancer 52:281–290
Translational Research in Lung Cancer
Deepinder Singh, Kevin Bylund, and Yuhchyau Chen

Contents Abstract
Translational research has become an important
1 Introduction.............................................................. 794 initiative emphasized by the National Institute of
2 Tumor Markers of Non-small Health (NIH) in recent years, with goals of
Cell Lung Cancer .................................................... 794 bringing benchtop research to the bedside through
2.1 Molecular Markers of Prognosis............................... 794 clinical trials, and translating clinical trials to the
2.2 Predictive Tumor Markers and Molecular
recommended care in clinical practice. Much
Targets........................................................................ 796
progress has been made in the translational
3 Small Cell Lung Cancer Serum Markers ............ 801 research of lung cancer. Molecular predictive
3.1 Chromogranin A ........................................................ 801
3.2 Neuron-Specific Enolase ........................................... 801 tumor markers, such as mutation of epidermal
3.3 Pro-Gastrin-Releasing Peptide .................................. 801 growth factor receptor (EGFR), markers of DNA
repair (excision repair cross-complementing group
4 A Chemoradiation Model of Translational
Investigation for Stage III Non-Small Cell 1 gene product [ERCC1]; regulator subunit of
Lung Cancer............................................................. 802 ribonucleotide reductase [RRM1]; human MutS
4.1 National Institutes of Health Roadmap homologue 2 [hMSH2], human MutL homologue
for Translational Medicine........................................ 802
1 [hMLH1]), and echinoderm microtubule-associ-
4.2 Translational Investigation Model of Taxane
Chemoradiation for NSCLC ..................................... 802 ated protein-like 4-anaplastic lymphoma kinase
(EML4-ALK) have been identified to predict
5 Summary................................................................... 804
response to molecular targeting agents for non-
References.......................................................................... 805 small cell lung cancer (NSCLC). Serum proteomic
profiling was found to predict response to EGFR
tyrosine kinase inhibitor (TKI) treatment of
NSCLC. Serum markers such as chromogranin
A (CgA), neuron-specific enolase (NSE), and pro-
gastric-releasing peptide (ProGRP) may aid diag-
nosis, detection of disease progression or recur-
rence, and monitoring therapy for small cell lung
cancer (SCLC). We present two clinical trial
designs that are based on preclinical investigations
that offer the rationale and hypotheses for taxane-
based chemoradiation treatment to maximize chest
D. Singh  K. Bylund  Y. Chen (&) tumor control and to target distant micrometastasis
University of Rochester Medical Center,
601 Elmwood Ave, Box 647, for stage III NSCLC. While stage III NSCLC in
Rochester, NY 14642, USA general is associated with[50% chest failure and a
e-mail: Yuhchyau_Chen@urmc.rochester.edu

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 793
DOI: 10.1007/174_2011_296, Ó Springer-Verlag Berlin Heidelberg 2011
794 D. Singh et al.

median survival of 14–17 months after chemora- individually, but lost the values when multiple factors
diation treatments, the outcome of this transla- were examined at the same time (Chen and Gandara
tional approach has yielded a 97% in-field tumor 2006). Here, we summarize and update K-ras and
control in study U1597, and 32 months overall p53, the two major prognostic molecular markers for
survival in study U1500. NSCLC, as well as review the current state of gene
signature prognostic factors in NSCLC.

1 Introduction 2.1 Molecular Markers of Prognosis

Translational research for lung cancer is evolving. 2.1.1 Ras Oncogenes p21
Progress has been made concerning molecular markers The ras oncogene family encodes guanosine triphos-
of lung cancer, which include tumor genetic markers, phate-binding proteins with a 21 kD (p21) molecular
tumor protein markers, and secreted proteins in the weight. The proteins are localized at the inner surface
serum of lung cancer patients. Molecular markers can of the cell membrane and are involved in the trans-
provide the prognosis of tumors expressing such mark- duction of growth signals. There are three well-
ers, or predict the response to treatment for certain types characterized members of the ras oncogene family:
of therapy. The latter is often associated with therapeutic H-ras, K-ras, and N-ras (Barbacid 1987). The onco-
agents that directly target the molecular markers within genic potential of ras genes is triggered by point
the tumor cells. The progress and success in molecular mutations occurring mainly in either codon 12, 13, or
targeting have formed the basis of many new drug 61. A ras gene mutation is detected in 10–30% of
developments by pharmaceutical companies. This NSCLC cases, and 80–90% of ras mutations occurred
approach has proven to be effective in ‘‘personalized at codon 12 of the K-ras gene (Rodenhuis et al. 1987;
medicine’’ regarding lung cancer treatment in recent Bos 1989; Slebos et al. 1990; Mitsudomi et al. 1991;
years. We summarize the current state of translational Sugio et al. 1992). The mutations are frequently
research in molecular markers of lung cancer and observed in smokers, are more frequent in adenocar-
describe a model in translational chemoradiation treat- cinoma than in squamous cell carcinoma (SCC), and
ment of locally advanced non-small cell lung cancer are absent in small-cell lung cancer (SCLC)
(NSCLC) in the context of the National Institutes of (Rodenhuis et al. 1987; Mitsudomi et al. 1991;
Health (NIH) roadmap with the translational emphasis Greatens et al. 1998; Brose et al. 2002; Riely et al.
from bench top to clinic (Westfall et al. 2007) (Fig. 1). 2009). Several investigators have reported that K-ras
mutations are a poor prognostic factor in NSCLC, and
the absence of H-ras p21 expression was among the
2 Tumor Markers of Non-small Cell nine independent predictors for recurrence, while
Lung Cancer other researchers found no negative prognostic value
to the K-ras mutation. More recently, Mascaux et al.
Numerous molecular markers for NSCLC have been (2005) performed a meta-analysis of more than 53
investigated in recent decades, including epidermal studies, which evaluated the K-ras mutation and
growth factor receptor (EGFR) family (c-erb-B1, outcomes in patients with NSCLC. They identified K-
c-erb-B2, c-erb-B3 and c-erb-B4), CD82, Ki-67, ras mutations or p21 overexpression by PCR assay as
p120, p53, bcl-2, CD31 MIA-15–5, p21, PCNA, a negative prognostic factor for NSCLC (hazard ratio
Ki-67, p185new protein, RB protein, blood group A, [HR] for death was 1.35; 95% confidence interval
K-ras mutation, K-ras p21 protein, H-ras p21 protein, [CI], 1.16–1.56), and even worse HR for adenocar-
angiogenic marker factor viii, adhesion molecule cinoma (HR 1.40; 95% CI 1.18–1.65). Notably,
CD-44, sialyl-Tn, blood group AMRP-1/CD9 gene, investigations of ras using immunohistochemical
KA11/CD82, laminin receptor, FOS, JUN, cyclin A, stain (IHC) showed no prognostic values in the meta-
TGF-a, amphiregulin (AR), and others (Chen and analysis (HR 1.08; 95% CI 0.86–1.34). The major
Gandara 2006). Some molecular markers were criticism of such meta-analysis was that it was not
found to have prognostic values when investigated based on prospectively designed studies, and the
Translational Research in Lung Cancer 795

Fig. 1 The National Institutes of Health (NIH) roadmap for (blue) includes an additional research arena (Practice-based
medical research includes two major research arenas (bench research) and translational step (T3) to improve incorporation
and bedside) and two translational steps (T1 and T2). Histor- of research discoveries into day-to-day clinical care. The
ically, moving new medical discoveries into clinical practice research roadmap is a continuum, with overlap between sites of
(T2) has been haphazard, occurring largely through continuing research and translational steps (Westfall et al. 2007, with
medical education programs, pharmaceutical detailing, and permission)
guideline development. The expansion of the NIH Roadmap

authors did not perform multivariate analyses to (P = 0.09). In multivariate analysis, the K-ras
include other clinical prognostic variables such as mutation was not an independent prognostic factor,
tumor stage, performance status, and weight loss. suggesting that it did not carry a distinct prognosis in
A large trial that has prospectively assessed K-ras this population of resected NSCLC.
mutations was conducted as part of the Eastern
Cooperative Oncology Group study E3590. Patients 2.1.2 p53 Tumor Suppressor Gene
with stage II–IIIA NSCLC were randomized to p53 is a tumor suppressor gene (TSG) encoding a
receive postoperative radiation therapy or radiation 53 kD nuclear phosphoprotein with a transcriptional
therapy and chemotherapy (Schiller et al. 2001). The activator, which controls cell proliferation by regulat-
study found no correlation of survival or disease-free ing a G1-S checkpoint before DNA synthesis, through
survival with K-ras mutations. Tumors from 197 the cyclin- dependent kinase (CDK) pathway (Cordon-
patients were available for K-ras mutational analysis Cardo 1995). In response to DNA damage by ionizing
out of the 488 patients enrolled. Mutations were radiation and a variety of chemical agents or carcino-
identified in 24% (44/184 cases), with 4.8% (3/63 gens, p53 induces cells to repair damage or promote
cases) in SCC and 33% in non-SCC histology apoptosis (Yin et al. 1992; Hartwell 1992; Farmer et al.
(P \ 0.05). Median survival was 30 months among 1992). Mutations of the p53 gene are the most common
the 44 patients with the K-ras mutation (95% CI: findings in human cancer cells of all types (Greenblatt
34–64 months), and 42 months among the 140 et al. 1994). The p53 gene is the most commonly
patients with wild type K-ras (P = 0.38). There was a mutated TSG in human malignancies, and these affect
trend toward significance for patients on the chemo- 90% of SCLC and 50% of NSCLC (Mayne et al. 1999).
radiation arm of the study. Seventy patients on the Most mutations occur in evolutionary conserved p53
chemoradiation arm who had wild type K-ras exon 5–8. p53 mutations correlate with smoking and
had a median survival of 42 months, compared with most are of the type G to T transversions expected from
25 months among the 20 patients with K-ras mutations tobacco smoke carcinogens.
796 D. Singh et al.

More evidence linking smoking damage with p53 2.1.3 Gene Expression Profiles
is that a major cigarette-smoking carcinogen known Gene expression profiling of resected lung tumors was
as benzopyrene selectively forms adducts at the p53 made possible by microarray technologies and real-
mutation hot spot (Kohno and Yokota 1999). The time reverse-transcriptase polymerase chain reaction
types of p53 mutations are varied and include mis- (RT-PCR) analysis. Gene expression profiles have
sense, nonsense, and splicing abnormalities as well as revealed the underlying heterogeneity of NSCLC
larger deletions. The most common mutations are of classified by conventional histopathology, thus
the missense type, which often prolongs the half-life offering the possibility of molecular taxonomy
of the p53 protein to several hours, leading to (Bhattacharjee et al. 2001). Further, several studies
increased levels detectable by IHC, which can be used have reported the prognostic value of discrete gene
for surrogate assay of p53. It is still controversial signatures in association with survival outcome for
whether these mutations detected in p53 affect operable NSCLC. These gene expression profiles
survival. were generated from resected NSCLC with mostly
The prognostic significance of p53 remains unclear stage I tumors. Prognostic gene signatures included
as studies have showed conflicting findings. There are 3–6 genes (Lau et al. 2007; Chen et al. 2007; Boutros
at least three published works on p53 meta-analyses. et al. 2008), 20–64 genes (Guo et al. 2008; Lu et al.
Mitsudomi et al. (2000) performed a meta-analysis of 2006; Sun et al. 2008), 125 genes (Larsen et al. 2007),
43 articles. p53 alteration was detected either by and more than 2,000 genes in the scenario of
overexpression of the protein (IHC) or as mutation by metagenes (Potti et al. 2006). All of these gene
the DNA studies. They found that the incidence of signatures have shown prognostic value for cancer
p53 alteration in DNA studies was 37% (381/1,031) survival independently or in combination with clinical
and the incidence of protein overexpression was 48% factors. The largest gene signature set is the meta-
(1,725/3,579 cases). The incidence of p53 overex- gene, which is a collection of gene expression profiles
pression and mutation in adenocarcinoma (36 and that are computer generated and randomly assigned
34%, respectively) was lower than that in SCC (54 sets of 25–200 genes. The signatures contain 100
and 52%, respectively). p53 alteration had a signifi- metagenes, which encompass more than 2,000 dis-
cant negative prognostic effect for adenocarcinoma tinct genes. In theory, this large size allows for a
but not for SCC. They concluded that p53 alteration much greater predictive power for disease recurrence
either by protein overexpression or by DNA mutation than fewer numbers of genes. Initially it was reported
was a significant marker of poor prognosis in patients with an accuracy of 72–93% in different patient
with pulmonary adenocarcinoma. Steels et al. (2001) groups with stage IA NSCLC (Potti et al. 2006).
did a meta-analysis of 74 eligible papers. The studies However, the original report was retracted in March
were categorized by histology, disease stage, treat- 2011 due to failure to reproduce results supporting the
ment, and laboratory technique. Combined hazard validation of the lung metagene model described in
ratios suggested that an abnormal p53 status had an the article by Potti et al. (2011). Nonetheless, the
unfavorable impact on survival for all tumor stages prognostic value of global gene profiling remains an
(I–IV) and for both SCLC and adenocarcinoma. area of active research interest.
Huncharek et al. (2000) published a meta-analysis of
eight studies investigating p53 mutations involving a
total of 829 patients. They did not find the p53 2.2 Predictive Tumor Markers
mutation as a prognostic marker in NSCLC, and felt and Molecular Targets
that selection bias, smoking history, race, geographic
location of the study, and socioeconomic status might 2.2.1 Epidermal Growth Factor Receptor
have been the confounding factors. Testing the Proto-oncogenes: c-erb-B1
prognostic value of p53 in a prospective study by the c-erb-B1 (EGFR) is the proto-oncogene encoding the
above-mentioned large trial of E3590, the study did EGFR protein. EGFR is a member of the erb-B family
not find the correlation of survival or disease-free of tyrosine kinase receptor proteins, which also
survival with p53 protein expression and p53 muta- include erb-B2 (HER-2/neu), erb-B3, and erb-B4
tion (Schiller et al. 2001). (Franklin et al. 2002). Intracellular signaling is
Translational Research in Lung Cancer 797

Fig. 2 Schematic
presentation of epidermal
growth factor receptor
(EGFR) activation. On
extracellular ligand (EGF)
binding, the receptor
dimerizes, allowing the
cytoplasmic EGFR-tyrosine
kinase (TK) to activate in a
tail-to-head fashion. The
locations of regions within
EGFR-TK are indicated on
the exon boundary map
(Kumar et al. 2008, with
permission)

triggered by the binding of ligands, such as epidermal number had any significant influence as an indepen-
growth factor (EGF), resulting in the dimerization dent prognostic factor. Poor prognosis was observed
of EGFR molecules or heterodimerization with in tumors with high gene copy numbers combined
other closely related receptors, such as HER-2/neu. with a low EGFR score by IHC. There is no data to
Phosphorylation of the receptors through their tyro- support or confirm whether high EGFR gene copy
sine kinase domains leads to intracellular signal number or overexpression correlates with survival
transduction and the activation of proliferative signals improvement after EGFR inhibitor therapy, thus
and DNA synthesis (Fig. 2) (Yarden and Sliwkowski such information is not useful in routine practice
2001; Jorissen et al. 2003; Kumar et al. 2008). In (Ramalingham et al. 2011).
NSCLC, EGFR is more commonly overexpressed In contrast to the lack of prognostic and predictive
than HER-2/neu, and has been observed in 40–80% of value of protein overexpression and gene copy num-
cancer specimens (Franklin et al. 2002; Arteaga 2003; ber, the somatic mutation of EGFR gene in the tyro-
Berger et al. 1987). The prognostic value of EGFR sine kinase domain was found to be predictive for
overexpression analyzed by IHC stains and by fluo- tumor response to treatment by EGFR tyrosine kinase
rescence in situ hybridization (FISH) in lung cancer inhibitors (TKIs): gefitinib (Iressa) and erlotinib
has been a controversial issue. Some reports indicated (Tarceva). EGFR mutations are present in approxi-
that EGFR overexpression was associated with a poor mately 10–15% of Caucasians and in nearly 40% of
prognosis (Volm et al. 1992; Ohsaki et al. 2000; Cox Asians. Patients with EGFR mutations are highly
et al. 2000), while others have shown no prognostic responsive to EGFR TKI treatment. Two early studies
association (Greatens et al. 1998; Rusch et al. 1997; support the concept of molecular targeting to specific
Pfeiffer et al. 1996; Fontanini et al. 1998; D’Amico et markers in predicting tumor responses, but did not
al. 1999; Pastorino et al. 1997). Hirsch et al. (2003) address the impact on survival. Lynch et al. (2004)
has reported analyses of the gene copy number of found that a subgroup of patients who had mutations
EGFR using the FISH technique as well as the protein in the EGFR gene by either in-frame deletions or
expression using IHC stain. In this report, EGFR amino acid substitutions around the ATP-binding
protein overexpression was observed in 62% of pocket of the tyrosine kinase domain had clinical
NSCLC with more frequency in SCC (82%) than non- responsiveness to gefitinib. Likewise, Paez et al.
SCC (44%), and in 80% of the bronchioloalveolar (2004) found somatic mutation in the kinase domain
carcinomas. They found that EGFR overexpression (exons 18 through 24) in 5/5 patients who responded
correlated with increased gene copy number per cell, to gefitinib, and none in four patients who did not
but neither EGFR overexpression nor high gene copy respond to gefitinib. Most of the patients with the
798 D. Singh et al.

mutations were women, non-smokers, and those with assigned to receive gefitinib or cisplatin plus docetaxel
bronchioloalveolar tumors. Somatic mutations of for 3–6 cycles. The gefitinib group had significantly
EGFR were more frequent in adenocarcinomas (21%) longer progression-free survival compared with the
than other NSCLC (2%), more frequent in women cisplatin plus docetaxel group, with a median pro-
(20%) than in men (9%), and more frequent in gression-free survival time of 9.2 months (95% CI
patients from Japan (26%) than patients from the US 8.0–13.9) versus 6.3 months (range 5.8–7.8; HR 0.489,
(2%). The highest fraction of EGFR mutations was 95% CI 0.336–0.710, log-rank P \ 0.0001). Again, it
found in Japanese women with adenocarcinomas was found that patients with NSCLC who were selected
(57%) (Kris et al. 2003; Fukuoka et al. 2003). by EGFR mutations had longer progression-free sur-
The benefit of EGFR TKIs for NSCLC patients has vival if they were treated with gefitinib than if they were
subsequently been established in randomized clinical treated with cisplatin plus docetaxel. Maemondo et al.
trials using erlotinib versus placebo in patients that (2010) randomly assigned 230 patients with metastatic
have progressed after 1 or 2 cycles of chemotherapy NSCLC and EGFR mutations without prior chemo-
for advanced-stage NSCLC. The NCI Canada BR21 therapy to receive gefitinib or carboplatin plus paclit-
trial found a significant improvement in overall sur- axel. In the planned interim analysis of the first 200
vival and progression free survival with erlotinib patients, progression-free survival was significantly
treatment, leading to FDA approval for the treatment longer in the gefitinib group than in the standard-
of refractory NSCLC (Brown and Shepherd 2005). chemotherapy group (HR for gefitinib group, 0.36;
A similar study was carried out with gefitinib, but this P \ 0.001), resulting in early termination of the study.
revealed no difference in overall survival when The gefitinib group had a significantly longer median
compared with placebo treatment (Thatcher et al. progression-free survival than the chemotherapy group
2005). However, in this latter study, significant sur- (10.8 vs. 5.4 months) (HR 0.30; 95% CI 0.22–0.41;
vival benefit was noted for patients who have never P \ 0.001), as well as a higher response rate (73.7 vs.
smoked and patients of Asian ethnicity treated with 30.7%, P \ 0.001). The median overall survival was
gefitinib in a subset analysis. 30.5 months in the gefitinib group and 23.6 months in
Recent large studies conducted in Asia and Spain the chemotherapy group (P = 0.31).
further confirmed the role of EGFR mutations as a
major predictor of outcome to treatment by EGFR 2.2.2 Markers of DNA Repair: Excision
TKIs (Rosell et al. 2009; Mok et al. 2009; Lee et al. Repair Cross-Complementing Group 1
2009; Mitsudomi et al. 2010; Maemondo et al. 2010). Gene Product and Regulatory Subunit
Findings from these large studies led to the paradigm of Ribonucleotide Reductase
shift in using EGFR TKIs for first line therapy in The excision repair cross-complementing group 1
patients with advanced stage NSCLC, with the benefit gene product (ERCC1) and the regulatory subunit of
limited to only patients with known EGFR mutations. ribonucleotide reductase (RRM1) have been reported
The study by Mok et al. (2009) was a randomized as being prognostic of outcome and predictors of
phase 3 study for those who had advanced pulmonary therapeutic efficacy in patients with NSCLC. Both
adenocarcinoma, and who were non-smokers or for- ERCC1 and RRM1 are critical to the nucleotide
mer light smokers to receive gefitinib versus carbo- excision repair (NER) of the DNA repair pathway.
platin plus paclitaxel chemotherapy in East Asia. Expression of these genes can both protect the host
Twelve-month rates of progression-free survival were from the development or progression of cancer, and
24.9% in the gefitinib arm, and 6.7% in the carbo- protect tumors from the effects of chemotherapy. Low
platin–paclitaxel arm. The study showed superiority ERCC mRNA expression and low RRM1 mRNA
of gefitinib in the intention-to-treat population (HR expression have been correlated with improved sur-
0.74; 95% CI: 0.65-0.85; P \ 0.001). Likewise, vival in advanced NSCLC after treatment with cis-
Mitsudomi et al. (2010) conducted a phase 3 study platin-based chemotherapy and decreased survival in
(WJTOG3405) in the Japanese population for early stage NSCLC with no adjuvant treatment
stage IIIB/IV NSCLC or postoperative recurrence (Rosell et al. 2004; Simon et al. 2008; Bepler et al.
harboring EGFR mutations. Patients were randomly 2008; Souglakos et al. 2008; Zheng et al. 2007).
Translational Research in Lung Cancer 799

2.2.3 Excision Repair Cross-Complementing prospective randomized clinical trial testing the con-
Group 1 Gene Product cept of customized chemotherapy in NSCLC has been
ERCC1 encodes a DNA-repair protein, a member of completed, which assessed the role of ERCC1 in
the NER complex. In particular, the ERCC1 protein treatment planning. The phase 3 study had 346
forms a heterodimer with xeroderma pigmentosum patients with stage IV NSCLC and they were evalu-
(XPF) group A protein, and functions to create the 50 ated after being randomized to receive either doce-
incision of damaged DNA. It is a highly conserved taxel and cisplatin or a tailored regimen based on
protein, and plays a rate-limiting role in the NER ERCC1 mRNA levels (docetaxel and cisplatin for low
pathway. ERCC1 polymorphisms have been impli- ERCC1 levels, and docetaxel plus gemcitibine for
cated in carcinogenesis (Chen et al. 1998). However, higher ERCC1 levels). A significant improvement in
ERCC1 function can also predict the capacity of the objective response rate was seen in the arm
tumor cells to repair after cytotoxic treatment, as NER receiving the tailored treatment. However, there was
is the primary mechanism for removing platinum– no difference in disease-free survival and overall
DNA adducts from the tumor DNA (Reed 1998). survival in the study arms (Cobo et al. 2007).
ERCC1 function and the NER pathway have also
been shown in vitro to explain the synergy between 2.2.4 Ribonucleotide-Diphosphate
cisplatin and gemcitibine (Yang et al. 2000). Reductase M1
A retrospective study evaluated ERCC1 mRNA The ribonucleotide-diphosphate reductase M1
expression in 56 patients with advanced (stage IIIB or (RRM1) gene is the large regulatory subunit of ribo-
IV) NSCLC (Lord et al. 2002). These patients were nucleotide reductase, which catalyzes the production
treated with cisplatin and gemcitibine. The study of deoxyribonucleotides from ribonucleotides prepa-
demonstrated ERCC1 expression to be a significant ratory to DNA synthesis during the S phase of the cell
predictor of overall survival, with ERCC1 low- cycle. RRM1 is also essential for NER, as it provides
expressing patients surviving 61.6 weeks as compared the bases needed for restoration of the complimentary
to 20.4 weeks in the ERCC1 high-expressing group, DNA strand. RRM1 also plays a role in cell migration
although improved tumor response to chemotherapy and metastases. The gene is encoded on 11p15.5, and
could not be shown. Subsequent retrospective studies is thought to be a TSG. In fact, RRM1 transgenic
using cisplatin and gemcitibine have confirmed an mice were significantly less likely to develop car-
overall survival either singly (Ceppi et al. 2006; cinogen-induced lung tumors (Gautam and Bepler
Hwang et al. 2008; Li et al. 2010) or in combination 2006). It is the dominant molecular determinant of
with low RRM1 expression (Rosell et al. 2004). Other gemcitabine efficacy.
studies, which have used other cisplatin-based thera- A phase II study was performed in 53 patients with
pies, have shown no increase in overall survival with previously untreated NSCLC who were stratified in
low expression of ERCC1 (Booton et al. 2007; Fujii et treatment based on real-time quantitative PCR mRNA
al. 2008), although a differential tumor response was levels of ERCC1 and RRM1 (Simon et al. 2008).
seen (Fujii et al. 2008). Treatments by different combinations of double agent
The International Adjuvant Lung Cancer Trial chemotherapy were based on low level versus high
(IALCT), which randomized patients to receive cis- level of ERCC1 and RRM1, showing feasibility of
platin-based chemotherapy after a complete resection this approach. In contrast to the expression in
of stage I–III NSCLC, showed a small but statistically advanced stage lung cancer, low RRM1 and ERCC1
significant improvement in overall survival with the expression appears to predict worse clinical outcomes
addition of chemotherapy (44.5 vs. 40.4% at five in patients with earlier stage disease. A study of 187
years) (Arriagada et al. 2004). However, when this patients with early stage NSCLC who received no
study was retrospectively analyzed for ERCC1 pro- adjuvant therapy after surgery showed that RRM1 and
tein expression, it was found that only patients with ERCC1 were both determinants of survival (Zheng et
low ERCC1 expression derived benefit from the al. 2007). This apparent paradox may be explained by
adjuvant therapy, while low RRM1 scores trended to the fact that intact DNA repair mechanisms in early
indicate benefit from adjuvant chemotherapy stage lung cancer may either prevent or compromise
(Olaussen et al. 2006; Bepler et al. 2011). The first the gathering of further genomic mutations and
800 D. Singh et al.

progression of disease. Thus, ERCC1 and RRM1 may 2.2.6 Human MutS Homologue 2 and
both predict patients with early stage tumors that do Human MutL Homologue 1
not require further therapy, and also inform appro- The human MutS homologue 2 (hMSH2), and human
priate chemotherapy for patients with more advanced MutL homologue 1 (hMLH1) genes are key compo-
cancers (Gazdar 2007). nents in DNA mismatch repair processes for the
recognition and replacement of erroneous base pairs.
2.2.5 Echinoderm Microtubule-Associated Mismatch repair reduces mutations at DNA replica-
Protein-Like 4-Anaplastic Lymphoma tions, but is also thought to be involved, as an adjunct
Kinase Translocation to NER, with error-free repair of DNA inter-strand
The echinoderm microtubule-associated protein-like crosslinks (Wu et al. 2005). Dysfunction of these
4-anaplastic lymphoma kinase (EML4-ALK) fusion- genes can lead to microsatellite instability, or errors in
type tyrosine kinase is an oncoprotein found in 4–5% of replicating repeating DNA sequences. Clinically,
NSCLCs. The EML4-ALK fusion gene is generated by mutations in these genes constitute hereditary non-
small inversion within the chromosome two short arm, polyposis colorectal cancer types 1 (for MSH2) or
encoding a 1,059-amino acid fusion protein. The type 2 (for MLH1), wherein patients carry a high risk
N-terminal portion is identical to the human EML4 for various gastrointestinal, genitourinary, and repro-
(Pollmann et al. 2006) and the C-terminal portion is the ductive system cancers.
same as the intracellular domain of human ALK (Morris Certain polymorphisms in the hMLH1 and hMSH2
et al. 1994). The EML4-ALK translocation is most genes have been shown to be associated with a
common in younger males, never smokers, and patients decreased or increased risk of developing NSCLC
with adenocarcinoma. EML4 NSCLC occurs most (Jung et al. 2006; Hsu et al. 2007; Kim et al. 2010; Lo
commonly in clinical subgroups, which share many of et al. 2011; Shih et al. 2010). In particular, decreased
the clinical features of NSCLC likely to harbor EGFR expression of the hMLH1 gene was seen more in
mutation (Shaw et al. 2009). With rare exceptions, EML SCLC, and decreased expression of the hMSH2
and EGFR mutations are mutually exclusive. EML was more common in adenocarcinoma of NSCLC
translocation tends to occur in younger patients and (Xinarianos et al. 2000). In early stage NSCLC, with
those with locally advanced disease, while this rela- no adjuvant therapy, MLH1 and MSH2 protein
tionship has not been reported for EGFR-mutant expression has not been shown to be prognostic
NSCLC (Inamura et al. 2009). There is currently no (Cooper et al. 2008). However, hMLH1 gene inacti-
standard method for detecting EML4 in NSCLC, and vation (Xinarianos et al. 2000), and in general,
several methods including PCR, IHC, and FISH are microsatellite instability (Woenckhaus et al. 2003) are
being evaluated. The challenge remains to incorporate associated with the risk of lymph node metastases.
and disseminate wide spread use of diagnostic testing for This indicates that mismatch repair defects can play a
identifying this patent subset (Sasaki et al. 2010). role in the development and progression of NSCLC.
As a molecular target, ALK inhibitors lead to The data are less clear, as in other DNA repair
apoptosis in vitro and tumor shrinkage in vivo. genes, as to whether inhibition of MSH2 correlates
Patients with tumor mutations may represent a very with improved survival in lung cancer patients
effective therapeutic strategy (Sasaki et al. 2010). requiring chemotherapy. A retrospective analysis of
ALK-targeted therapies are in advanced clinical the IALCT showed that patients with low MSH2
development of EML4-ALK NSCLC. PF-02341066 protein levels (about 38% of the whole) had trends
(crizotinib) is an orally bio available ALK inhibitor towards improved survival, and that no benefit was
currently under clinical development. Initial findings seen when MSH2 was high. Chemotherapy prolonged
demonstrated a remarkable 53% response rate and overall survival significantly for patients with tumors
disease control rate of 79% (Kwak et al. 2009). These expressing low levels of both MSH2 and ERCC1, and
dramatic findings have led to two clinical trials of also for low levels of MSH2 and p27 (a cell cycle
PF-023410066. A randomized phase 3 trial that protein) (Kamal et al. 2010). Another study of 179
compares PF-02341066 with standard second line patients did not show any association of hMLH1 or
chemo premetrexed or docetaxel in second line hMSH2 expression with disease outcomes in stage III
EMLALK NSCLC cancer patients is underway. NSCLC (Skarda et al. 2006). A third study showed
Translational Research in Lung Cancer 801

improved survival for cases with low expression of 3.1 Chromogranin A


hMLH1, but not hMSH2 (Scartozzi et al. 2006). Data to
date indicate that decreased activity of the mismatch CgA is a 49 kD acidic-soluble protein ubiquitously
repair pathway can be associated with development and present in neuroendocrine tissues and serves as a
progression of NSCLC. In more advanced disease, suitable circulating marker of neoplasms of neuro-
decreased activity in this pathway does not correlate endocrine origin. The ability of serum CgA to dis-
robustly with an increased response to cytotoxic tinguish neuroendocrine and non-neuroendocrine
chemotherapy. This may indicate that although such tumors either in situ or by serum level titration has
cells may have a decreased ability to repair chemo- been suggested (Drivsholm et al. 1999).
therapy-induced defects, this is balanced by a muta-
genic phenotype, allowing for phenotypic selection.
3.2 Neuron-Specific Enolase
2.2.7 Serum Proteomic Profiling
Serum proteomic profiling that utilizes the matrix- NSE is a neuronal form of the glycolytic enzyme
assisted laser desorption/ionization mass spectrometry enolase, which was first found in extracts of brain
(MALDI) reveals many different proteins in the blood tissue, and was later shown to be present in amine
at the time of sample collection. It has been shown precursor uptake and decarboxylation (APUD) cells
that serum proteomic profiles of blood samples from and neurons of the diffuse neuroendocrine system, but
NSCLC patients can predict which patients are more not in other peripheral cells. Neuroendocrine neo-
likely to benefit from EGFR TKI treatment (Meyer- plasias (APUDomas) (including islet-cell tumors,
son and Carbone 2005). Some clinical trials have pheochromocytomas, medullary thyroid carcinomas,
been conducted and validated this observation SCLC, and APUDomas of the gut, pancreas, and
(Amann et al. 2010; Taguchi et al. 2007). lung) reacted strongly with antisera to NSE (Tapia et
al. 1981).

3 Small Cell Lung Cancer Serum


Markers 3.3 Pro-Gastrin-Releasing Peptide

SCLC is characterized by rapid growth and the GRP is a gut hormone that is present in nerve fibers,
propensity for early metastases. It is known to be as well as in the brain and neuroendocrine cells in the
associated with neuroendocrine differentiation due to fetal lung (Drivsholm et al. 1999; Miyake et al. 1994).
its cell origin. SCLC has shown positive expressions It was originally isolated from the porcine stomach
by IHC stain with neuroendocrine markers such as and is the mammalian counterpart of amphibian
Neuron Specific Enolase (NSE, an isoform of the bombesin. It is elevated in the plasma of patients with
ubiquitous enolase enzyme), chromogranin A (CgA), SCLC, but its regular use as a diagnostic marker is not
synaptophysin, creatinine kinase BB, bombesin preferred because of its instability in the serum with a
(Gastrin-Releasing Peptide [GRP]), and neural cell half-life of approximately 2 min (Yamaguchi et al.
adhesion molecule (NCAM) (Berendsen et al. 1989; 1983). In contrast, serum ProGRP, a more stable
Erisman et al. 1982; Yamaguchi et al. 1983). Some of precursor of GRP, has been shown to be a specific
these neuroendocrine molecules are released into the tumor marker of SCLC (Miyake et al. 1994).
blood of SCLC patients. Translational investigation of While all three markers are useful in the diagnostic
SCLC has been focused on these serum markers. At performance of SCLC, various studies have shown
present, SCLC serum markers are considered more that serum ProGRP is superior to other markers in its
useful for diagnosis, detection of disease progression ability to differentiate SCLC and NSCLC. Studies
or recurrence, and monitoring of therapy, but less also found that serum CgA has better diagnostic
useful for the prognosis or prediction of response to sensitivity than NSE in SCLC (61 vs. 57%), espe-
therapy for SCLC. The serum markers with more cially in limited disease. In contrast, NSE reflected
specificity to SCLC are CgA, NSE, and Pro-gastrin- disease extent more accurately than CgA. It has also
releasing peptide (Pro-GRP). been shown that the CgA assay is not affected by
802 D. Singh et al.

hemolysis, whereas NSE serum levels greatly trials in the investigation of taxane-based chemoradi-
increased in hemolysed sera. CgA assays via this ation treatment for stage III NSCLC. This project has
method are a reliable procedure in the diagnosis of discovered the differential effects between paclitaxel
SCLC, whereas NSE is the suitable marker of choice and docetaxel in the ‘‘radiosensitizing effect’’ versus
in staging and monitoring of the disease (Holdenrie- the ‘‘cytotoxic effect’’, and has applied these differen-
der et al. 2008; Wójcik et al. 2008). In a large retro- tials to maximally target the chest tumor control and
spective study examining the single and combined distant micrometastasis control of stage III NSCLC.
diagnostic value of NSE, ProGRP, serum carcino-
embryonic antigen (CEA), and cytokeratin fragment
19 (CYFRA21-1), ProGRP reached the highest diag- 4.2 Translational Investigation Model
nostic efficacy for SCLC with 57% true positive of Taxane Chemoradiation for NSCLC
results at a specificity of [99% (Gruber et al. 2008).
Serum markers have also been used for monitoring Locally advanced stage III NSCLC constitutes
first-line chemotherapy in the management of SCLC 20–30% of patients who present with NSCLC.
(Holdenrieder et al. 2008). A study assessing Pro-GRP, Treatment of stage III NSCLC remains a challenge to
NSE, CYFRA 21-1 and lactate dehydrogenase (LDH) oncologists. Despite aggressive chemoradiation
levels at the time of diagnosis and during chemotherapy combination treatment, the outcome is quite poor with
and radiotherapy of SCLC patients with limited stage of a high intrathoracic failure rate at approximately 50%
disease, found elevated levels of ProGRP, NSE, CYFRA and a high rate of distant metastasis at approximately
21-1, and LDH in 79.9, 57.8, 23.4, and 12.5% of patients, 80% (Morton et al. 1991; Schaake-Koning et al. 1992;
respectively. Before the second chemotherapy course, Mattson et al. 1988; Gandara et al. 1991; Marino et al.
all tumor marker levels except LDH decreased signifi- 1995; Arriagada et al. 1991; Sause et al. 2000; Curran
cantly in comparison with the pretreatment levels. et al. 2000; Furuse et al. 1999, 2000). The median
However, only ProGRP levels showed a progressive survival time using cisplatin-based chemoradiation is
decrease during consecutive course of therapy, while only 14–17 months from large randomized trials of
NSE and DYFRA 21-1 fluctuated within reference chemoradiation treatments. To improve the local
ranges. Changes of Pro-GRP level seem to be more tumor control rate, the Radiation Therapy Oncology
precise than NSE as a tool for monitoring therapy in Group (RTOG) is currently conducting a four-arm
SCLC patients with limited disease (Wójcik et al. 2008). phase III randomized clinical trial (RTOG 0617)
using concurrent chemotherapy (carboplatin/paclit-
axel), with or without cetuximab (monoclonal anti-
4 A Chemoradiation Model body against EGFR), and radiation, to compare
of Translational Investigation standard dose radiation of 60 Gy versus high dose
for Stage III Non-Small Cell Lung radiation to 74 Gy. In addition to adding molecular
Cancer targeted agents such as cetuximab to improve on the
control of distant metastasis, many other clinical trials
4.1 National Institutes of Health have applied newer chemotherapeutic agents and
Roadmap for Translational Medicine tested different chemoradiation sequences. Thus far,
there is no major therapeutic breakthrough in
For decades, the NIH has been funding basic biomed- improving the treatment outcome of stage III NSCLC
ical research that aims to understand how living (Chen and Okunieff 2004).
organisms work. The initiative of the NIH roadmap A translational investigation was conducted at the
(Fig. 1), has increased the focus on the need to University of Rochester by bringing preclinical
‘‘translate’’ basic research more quickly into human benchtop findings to the development of two
studies, and then into tests and treatments that can sequential clinical trials using taxane-base chemora-
improve clinical practice for the benefit of patients diation regimens. The two studies targeted chest
(Westfall et al. 2007). Here we describe a translational tumor control and distant micrometastasis control,
investigation that has successfully brought the yielding promising results (Chen et al. 2001, 2003,
‘‘benchtop’’ observations to the ‘‘bedside’’ of clinical 2010).
Translational Research in Lung Cancer 803

Fig. 3 Clonogenic survival curves of human lung cancer cell cell death (steeper slopes) if radiation is delayed at 24 h,
line NCI520. Cells were treated with either a paclitaxel (Taxol) supporting better radiosensitizing effect by delaying radiation
or b docetaxel (Taxotere) at two different drug concentrations after drug treatments. For sham irradiated cells (0 Gy), there is
(50 or 100 nM) for 3 h. After drug treatments, cells were either more cell death in the docetaxel treated cultures (Y axis of B)
irradiated immediately or irradiated 24 h later. Radiation doses than the paclitaxel treated cultures (Y axis of A), demonstrating
were 2, 4, 6, and 8 Gy versus sham radiation (0 Gy). Both that docetaxel is significantly more cytotoxic (Chen et al. 2001,
paclitaxel treated cells and docetaxel treated cells showed more 2010)

4.2.1 Preclinical Studies Of Lung Cancer Cell based chemotherapy and radiation therapy for the
Lines treatment of stage III NSCLC. Based on preclinical
Taxanes are the ideal radiation sensitizers due to the information regarding the cell cycle and apoptotic
cell cycle effect in arresting cancer cells in the most effects of paclitaxel on lung cancer cell lines, U1597
sensitive phase of the cell cycle of G2/M (Sinclair applied concurrent pulsed low-dose, radiosensitizing
1966). Investigators at the University of Rochester paclitaxel chemoradiation to treat stage III NSCLC.
conducted preclinical studies using lung cancer cell To optimize radiosensitization by paclitaxel without
lines and investigated effects of paclitaxel and doce- increasing toxicity, a chemoradiation combination
taxel on the cytotoxic effects versus radiation sensi- was conducted in a schedule dependent manner with
tizing effects of these taxanes. The preclinical data paclitaxel delivered on alternating days (three times
revealed the following: (i) after paclitaxel treatment, a per week, M, W, F) at low doses (15, 20, and 25 mg/
minimum of 4 h is necessary for cell cycle progression m2 in the phase I dose-escalation study). Daily radi-
to G2/M phase of the cell cycle, the most radiosensitive ation was delayed to allow for cell-cycle progression
phase of the cell cycle (Chen et al. 2001); (ii) pulsed of cancer cells into G2/M phase of the cell cycle, the
treatment of taxane sustains G2/M cell cycle arrest most radiosensitive phase of the cell cycle. The
(Chen et al. 2003); (iii) delaying radiation after taxane pulsed low-dose paclitaxel schedule allowed for rapid
treatment led to better radiosensitizing effects than clearance of drug from the plasma to minimize tox-
immediate radiation after drug treatment (Chen et al. icity (Chen et al. 2008). This phase I study (U1597)
2001, 2003, 2010), and (iv) docetaxel demonstrates a yielded a 100% gross tumor response rate, a three-
much more potent cytotoxic effect than paclitaxel year survival of 30%, and low rates of grade 3 or 4
(Chen et al. 2001, 2010) (Fig. 3). toxicities (Chen et al. 2001, 2003, 2008). The inves-
tigators contoured the tumors on 104 pre-treatment
4.2.2 Translating Preclinical Studies to and post-treatment chest CT scans, and modeled the
Clinical Trials kinetics of clinical tumor response (Zhang et al. 2008).
Translating the preclinical information to the design Data showed that the bulk of the tumor shrinkage
of two sequential clinical studies (U1597 and U1500) occurred within 4–6 weeks post-treatment, and larger
led to novel approaches in the combination of taxane- tumors shrunk faster (Fig. 4). This is dramatically
804 D. Singh et al.

Fig. 4 The tumor response kinetics model is developed from are shown in the table insert. The volume reduction over time
104 chest CTs of patients treated on U1597, employing pulsed shows early rapid tumor shrinkage within the first 4–6 weeks.
low-dose paclitaxel (Taxol) chemoradiation treatment for stage b The rate of tumor shrinkage versus the initial tumor volume
III NSCLC. a Mathematic model of tumor response kinetics. correlation shows that larger tumors shrink faster for tumors
Estimates of the model parameters and their standard deviation treated by pulsed paclitaxel chemoradiation (unpublished data)

different from the expected kinetics of NSCLC in that Median survival was 32 months in U1500 and was
tumor shrinkage after chemoradiation is expected to be 11 months in U1597.
slow and may take several months to reach nadir. This The clinical outcome of these two studies support
study yielded an in-field (radiation port) tumor control that while the addition of one-cycle full-dose che-
of 97% at 5 years (Chen et al. 2003). motherapy of docetaxel/cisplatin improved survival
To target distant micrometastasis upfront, the of patients, pulsed low-dose radiosensitization
second study (U1500) employed one-cycle induction ‘‘paclitaxel’’ was more effective than ‘‘docetaxel’’ in
chemotherapy of docetaxel and cisplatin, followed by sensitizing radiation for gross chest tumor control.
low-dose, pulsed docetaxel chemoradiation. The
hypothesis of U1500 was that for targeting micro-
metastasis, a single cycle of full-dose chemotherapy 5 Summary
should suffice, while two or three cycles of induction
chemotherapy will delay local therapy. This is based The emphasis of the NIH roadmap has yielded
on thoughts by Goldie (1987) that ‘‘it can be stated increasing success in translating benchtop research to
that as a general biological principle that there are clinical trials. Translation research in NSCLC has
many compelling reasons why chemotherapy should revealed that most molecular prognostic factors tested
be directed at minimal tumor burdens.’’ One-cycle by IHC are not useful. After rigorous testing, K-ras
induction chemotherapy in targeting potential mutation is associated with poor prognosis, but its
distant micrometastasis instead of the conventional prognostic value has not been observed in a pro-
2–3 cycles would not delay chest local therapy using spective clinical study for patients with stage II and
concurrent radiosensitizing chemoradiation. There- IIIA NSCLC population (Schiller et al. 2001). On the
fore, U1500 was conducted with one cycle of cisplatin other hand, major progress has been made with
(75 mg/m2) and docetaxel (75 mg/m2) followed by molecular markers in predicting response to particular
low-dose sensitizing docetaxel (12.5 to 15 mg/m2, treatments, and in some cases survival outcome as
twice per week on Monday and Thursday), and con- well. Findings in the predictive molecular markers
current daily chest radiation to 64.8 Gy. The result such as EGFR mutations, ERCC1, RRM1, EML4-
showed an overall response rate of 69%, which was ALK translocations, hMSH2, and hMLH1, have been
lower than the pulsed paclitaxel study of U1597 applied to pharmaceutical targets for personalized
(100%), but had a much better survival rate than the therapy in molecular targeting approaches to the
U1597 study. The 3-year overall survival was 45% treatment of NSCLC. Serum proteomics have been
compared to 30% in U1597 (Chen et al. 2003, 2010). found to be predictive in the treatment response to
Translational Research in Lung Cancer 805

EGFR TKI in NSCLC, while serum markers such as Brose MS, Volpe P, Feldman M et al (2002) BRAF and RAS
CgA, NSE, and Pro-GRP have the potential value in mutations in human lung cancer and melanoma. Cancer Res
62:6997–7000
the diagnosis and monitoring therapy response for Brown ER, Shepherd FA (2005) Erlotinib in the treatment of
SCLC. The taxane-based chemoradiation model we non-small cell lung cancer. Expert Rev Anticancer Ther
presented here supports that the translational investi- 5:767–775
gation to the design of clinical trials that are Ceppi P, Volante M, Novello S et al (2006) ERCC1 and RRM1
gene expressions but not EGFR are predictive of shorter
hypothesis based and are built upon preclinical survival in advanced non-small-cell lung cancer treated with
findings may have higher probability of yielding the cisplatin and gemcitabine. Ann Oncol 17:1818–1825
desirable clinical outcome. Chen HY, Yu SL, Chen CH et al (2007) A five-gene signature
and clinical outcome in non-small-cell lung cancer. N Engl
J Med 356:11–20
Chen P, Wiencke J, Aldape K et al (1998) Association of an
References ERCC1 polymorphism with adult-onset glioma. Cancer
Epidemiol Biomark Prev 9:843–847
Chen Y, Gandara D (2006) Molecular staging of non-small cell
Amann JM, Lee JW, Roder H et al (2010) Genetic and lung cancer. In: Syrigos KN, Nutting C, Roussos C (eds)
proteomic features associated with survival after treatment Tumors of the chest: biology diagnosis and management.
with erlotinib in first-line therapy of non-small cell lung Springer, Heidelberg pp 159–176
cancer in Eastern Cooperative Oncology Group 3503. J Chen Y, Okunieff P (2004) Radiation and third generation
Thorac Oncol 5:169–178 chemotherapy. Hematol Oncol Clin N Am 18:55–80
Arriagada R, Bergman B, Dunant A et al (2004) Cisplatin- Chen Y, Pandya K, Feins R et al (2008) Toxicity profile and
based adjuvant chemotherapy in patients with completely pharmacokinetic study of a phase I low-dose schedule-
resected non-small-cell lung cancer. N Engl J Med 350: dependent radiosensitizing paclitaxel chemoradiation regi-
351–360 men for inoperable non-small cell lung cancer (NSCLC). Int
Arriagada R, le Chevalier T, Quoix E et al (1991) ASTRO J Radiat Oncol Biol Phys 71:407–413
plenary: effect of chemotherapy on locally advanced non- Chen Y, Pandya KJ, Hyrien O et al (2010) Preclinical and pilot
small cell lung carcinoma: a randomized study of 353 clinical study of docetaxel chemoradiation for stage III
patients. Int J Radiat Oncol Biol Phys 20:1183–1190 non-small cell lung cancer. Int J Radiat Oncol Biol Phys
Arteaga CL (2003) ErbB-targeted therapeutic approaches in (Epub ahead of print: 12 Aug, 2010)
human cancer. Exp Cell Res 284:122–130 Chen Y, Pandya K, Keng P et al (2001) Schedule dependent
Barbacid M (1987) Ras genes. Ann Rev Biochem 56:779–827 pulsed low-dose paclitaxel radiosensitization for thoracic
Berendsen HH, de Leij L, Poppema S et al (1989) Clinical malignancy. Am J Clin Oncol 24:432–437
characterization of non-small-cell lung cancer tumors Chen Y, Pandya K, Keng PC et al (2003) Phase I/II clinical trial
showing neuroendocrine differentiation features. J Clin using pulsed low-dose paclitaxel radiosensitization for
Oncol 7:1614–1620 thoracic malignancies: a therapeutic approach based on
Berger MS, Gullick WJ, Greenfield C et al (1987) Epidermal pre-clinical research of human lung cancer cells. Clin
growth factor receptors in lung tumours. J Pathol 152: Cancer Res 9:969–975
297–307 Cobo M, Isla D, Massuti B et al (2007) Customizing cisplatin
Bepler G, Olaussen KA, Vataire AL et al (2011) ERCC1 and based on quantitative excision repair cross-complementing
RRM1 in the international adjuvant lung trial by automated 1 mRNA expression: a phase III trial in non-small-cell lung
quantitative in situ analysis. Am J Pathol 178:69–78 cancer. J Clin Oncol 25:2747–2754
Bepler G, Sommers KE, Cantor A et al (2008) Clinical efficacy Cooper WA, Kohonen-Corish MR, Chan C et al (2008)
and predictive molecular markers of neoadjuvant gemcita- Prognostic significance of DNA repair proteins MLH1,
bine and pemetrexed in resectable non-small cell lung MSH2 and MGMT expression in non-small-cell lung cancer
cancer. J Thorac Oncol 3:1112–1118 and precursor lesions. Histopathology 52:613–622
Bhattacharjee A, Richards WG, Staunton J et al (2001) Cordon-Cardo C (1995) Mutations of cell cycle regulators.
Classification of human lung carcinomas by mRNA expres- Biological and clinical implications for human neoplasia.
sion profiling reveals distinct adenocarcinoma subclasses. Am J Pathol 147:545–560
Proc Natl Acad Sci USA 98:13790–13795 Cox G, Jones JL, O’Byrne KJ (2000) Matrix metalloproteinase
Booton R, Ward T, Ashcroft L et al (2007) ERCC1 mRNA 9 and the epidermal growth factor signal pathway in
expression is not associated with response and survival after operable non-small cell lung cancer. Clin Cancer Res
platinum-based chemotherapy regimens in advanced non- 6:2349–2355
small cell lung cancer. J Thorac Oncol 2:902–906 Curran WJ Jr, Scott C, Langer C et al (2000) Phase III
Bos JL (1989) Ras oncogenes in human cancer: a review. comparison of sequential vs. concurrent chemoradiation for
Cancer Res 49:4682–4689 PTS with unresected stage III non-small cell lung cancer
Boutros T, Chevet E, Metrakos P (2008) Mitogen-activated (NSCLC) initial report of radiation therapy Oncology Group
protein (MAP) kinase/MAP kinase phosphatase regulation: (RTOG) 9410. Proc Am Soc Clin Oncol 19:1891
roles in cell growth, death, and cancer. Pharmacol Rev D’Amico TA, Massey M, Herndon JE II et al (1999) A biologic
60:261–310 risk model for stage I lung cancer: immunohistochemical
806 D. Singh et al.

analysis of 408 patients with the use of ten molecular Hartwell L (1992) Defects in a cell cycle checkpoint may be
markers. J Thorac Cardio Surg 117:736–743 responsible for the genomic instability of cancer cells. Cell
Drivsholm L, Paloheimo LI, Osterlind K et al (1999) 71:543–546
Chromogranin A, a significant prognostic factor in small Hirsch FR, Varella-Garcia M, Bunn PA Jr et al (2003)
cell lung cancer. Br J Cancer 81:667–671 Epidermal growth factor receptor in non-small-cell lung
Erisman MD, Linnoila RI, Hernandez O et al (1982) Human carcinomas: correlation between gene copy number and
lung small-cell carcinoma contains bombesin. Proc Natl protein expression and impact on prognosis. J Clin Oncol
Acad Sci USA 79:2379–2383 21:3798–3807
Farmer G, Bargonetti J, Zhu H et al (1992) Wild-type p53 Holdenrieder S, von Pawel J, Dankelmann E et al (2008)
activates transcription in vitro. Nature 358:83–86 Nucleosomes, ProGRP, NSE, CYFRA 21-1, and CEA in
Fontanini G, De Laurentiis M, Vignati S et al (1998) Evaluation monitoring first-line chemotherapy of small cell lung
of epidermal growth factor-related growth factors and cancer. Clin Cancer Res 14:7813–7821
receptors and of neoangiogenesis in completely resected Hsu HS, Lee IH, Hsu WH et al (2007) Polymorphism in the
stage I–IIIA non-small-cell lung cancer: amphiregulin and hMSH2 gene (gISV12–6T [ C) is a prognostic factor in
microvessel count are independent prognostic indicators of non-small cell lung cancer. Lung Cancer 58:123–130
survival. Clin Cancer Res 4:241–249 Huncharek M, Kupelnick B, Geschwind JF et al (2000)
Franklin WA, Veve R, Hirsch FR et al (2002) Epidermal Prognostic significance of p53 mutations in non-small cell
growth factor receptor family in lung cancer and premalig- lung cancer: a meta-analysis of 829 cases from eight
nancy. Semin Oncol 29(1 suppl 4):3–14 published studies. Cancer Lett 153:219–226
Greatens TM, Niehans GA, Rubins JB et al (1998) Do Hwang IG, Ahn MJ, Park BB et al (2008) ERCC1 expression as
molecular markers predict survival in non-small-cell lung a prognostic marker in N2(+) nonsmall-cell lung cancer
cancer? Am J Respir Crit Care Med 157:1093–1097 patients treated with platinum-based neoadjuvant concur-
Fujii T, Toyooka S, Ichimura K et al (2008) ERCC1 protein rent chemoradiotherapy. Cancer 113:1379–1386
expression predicts the response of cisplatin-based neoad- Inamura K, Takeuchi K, Togashi Y et al (2009) EML4-ALK
juvant chemotherapy in non-small-cell lung cancer. Lung lung cancers are characterized by rare other mutations, a
Cancer 59:377–384 TTF-1 cell lineage, an acinar histology, and young onset.
Fukuoka M, Yano S, Giaccone G et al (2003) Multi-institutional Mod Pathol 22:508–515
randomized phase II trial of gefitinib for previously treated Jorissen RN, Walker F, Pouliot N et al (2003) Epidermal
patients with advanced non-small-cell lung cancer (The growth factor receptor: mechanisms of activation and
IDEAL 1 trial). J Clin Oncol 21:2237–2246 signalling. Exp Cell Res 284:31–53
Furuse K, Fukuoka M, Kawahara M et al (1999) Phase III study Jung CY, Choi JE, Park JM et al (2006) Polymorphisms in the
of concurrent versus sequential thoracic radiotherapy in hMSH2 gene and the risk of primary lung cancer. Cancer
combination with mitomycin, vindesine, and cisplatin in Epidemiol Biomarkers Prev 15:762–768
unresectable stage III non-small-cell lung cancer. J Clin Kamal NS, Soria JC, Mendiboure J et al (2010) MutS
Oncol 17:2692 homologue 2 and the long-term benefit of adjuvant chemo-
Furuse K, Hosoe S, Masuda N et al (2000) Impact of tumor therapy in lung cancer. Clin Cancer Res 16:1206–1215
control on survival in unresectable stage III non-small cell Kim M, Kang HG, Lee SY et al (2010) Comprehensive analysis
lung cancer (NSCLC) treated with concurrent thoracic of DNA repair gene polymorphisms and survival in patients
radiotherapy (TRT) and chemotherapy (CT). Proc Am Soc with early stage non-small-cell lung cancer. Cancer Sci
Clin Oncol 18(suppl 1) abstr 1893 101:2436–2442
Gandara DR, Valone FH, Perez EA et al (1991) Rapidly Kohno T, Yokota J (1999) How many tumor suppressor genes
alternating radiotherapy and high dose cisplatin chemother- are involved in human lung carcinogenesis? Carcinogenesis
apy in stage IIIB non-small cell lung cancer. results of a 20:1403
Phase I/II study. Int J Radiat Oncol Biol Phys 20:1047–1052 Kris MG, Natale RB, Herbst RS et al (2003) Efficacy of
Gautam A, Bepler G (2006) Suppression of lung tumor gefitinib, an inhibitor of the epidermal growth factor
formation by the regulatory subunit of ribonucleotide receptor tyrosine kinase, in symptomatic patients with
reductase. Cancer Res 66:6497–6502 non-small cell lung cancer: a randomized trial. JAMA
Gazdar AF (2007) DNA repair and survival in lung cancer––the 290:2149–2158
two faces of Janus. N Engl J Med 356:771–773 Kumar A, Petri ET, Halmos B et al (2008) Structure and
Goldie JH (1987) Scientific basis for adjuvant and primary clinical relevance of the epidermal growth factor receptor in
(neoadjuvant) chemotherapy. Semin Oncol 14:1–7 human cancer. J Clin Oncol 26:1742–1751
Greenblatt MS, Bennett WP, Hollstein M et al (1994) Larsen JE, Pavey SJ, Bowman R et al (2007) Gene expression
Mutations in the p53 tumor suppressor gene: clues to of lung squamous cell carcinoma reflects mode of lymph
cancer etiology and molecular pathogenesis. Cancer Res node involvement. Eur Respir J 30:21–25
54:4855–4878 Kwak EL, Camidge DR, Clark J et al (2009) Clinical activity
Gruber C, Hatz R, Reinmiedl J et al (2008) CEA, CYFRA observed in a phase I dose escalation trial of an oral c-met
21–1, NSE, and ProGRP in the diagnosis of lung cancer: a and ALK inhibitor, PF-02341066. J Clin Oncol 27:15s
multivariate approach. J Lab Med 32:361–371 Abstract 3509
Guo NL, Wan YW, Tosun K et al (2008) Confirmation of gene Lau SK, Boutros PC, Pintilie M et al (2007) Three-gene
expression-based prediction of survival in non-small cell prognostic classifier for early-stage non small-cell lung
lung cancer. Clin Cancer Res 14:8213–8220 cancer. J Clin Oncol 25:5562–5569
Translational Research in Lung Cancer 807

Lee JS, Park K, Kim S et al (2009) A randomized phase III Mok TS, Wu YL, Thongprasert S et al (2009) Gefitinib or
study of gefitinib versus stand ard chemotherapy (gemcit- carboplatin–paclitaxel in pulmonary adenocarcinoma. N
abine plus cisplatin) as a first-line treatment for never- Engl J Med 361:947–957
smokers with advanced or metastatic adenocarcinoma of the Morris SW, Kirstein MN, Valentine MB et al (1994) Fusion of
lung. J Thorac Oncol 4:S283–S284 Abstract PRS 4 a kinase gene, ALK, to a nucleolar protein gene, NPM, in
Li J, Li ZN, Yu LC et al (2010) Association of expression of MRP1, non-Hodgkin’s lymphoma. Science 263:1281–1284
BCRP, LRP and ERCC1 with outcome of patients with locally Morton RF, Jett JR, McGinnis WL et al (1991) Thoracic
advanced non-small cell lung cancer who received neoadju- radiation therapy alone compared with combined chemora-
vant chemotherapy. Lung Cancer 69:116–122 diotherapy for locally unresectable non-small cell lung
Lo YL, Hsiao CF, Jou YS et al (2011) Polymorphisms of cancer. Ann Int Med 115:681–686
MLH1 and MSH2 genes and the risk of lung cancer among Ohsaki Y, Tanno S, Fujita Y et al (2000) Epidermal growth
never smokers. Lung Cancer 72:280–286 factor receptor expression correlates with poor prognosis in
Lord RV, Brabender J, Gandara D et al (2002) Low ERCC1 non-small cell lung cancer patients with p53 overexpres-
expression correlates with prolonged survival after cisplatin sion. Oncol Rep 7:603–607
plus gemcitabine chemotherapy in non-small cell lung Olaussen KA, Dunant A, Fouret P et al (2006) DNA repair by
cancer. Clin Cancer Res 8:2286–2291 ERCC1 in non-small-cell lung cancer and cisplatin-based
Lu Y, Lemon W, Liu PY et al (2006) A gene expression adjuvant chemotherapy. N Engl J Med 355:983–991
signature predicts survival of patients with stage I non-small Paez JG, Janne PA, Lee JC et al (2004) EGFR mutations in
cell lung cancer. PLoS Med 3:e467 lung cancer: correlation with clinical response to gefitinib
Lynch TJ, Bell DW, Sordella R et al (2004) Activating therapy. Science 304:1497–1500
mutations in the epidermal growth factor receptor underly- Pastorino U, Andreola S, Tagliabue E et al (1997) Immuno-
ing responsiveness of non-small-cell lung cancer to gefiti- cytochemical markers in stage I lung cancer: relevance to
nib. N Engl J Med 350:2129–2139 prognosis. J Clin Oncol 15:2858–2865
Maemondo M, Inoue A, Kobayashi K et al (2010) Gefitinib or Pfeiffer P, Clausen PP, Andersen K et al (1996) Lack of
chemotherapy for nonsmall-cell lung cancer with mutated prognostic significance of epidermal growth factor receptor
EGFR. N Engl J Med 362:2380–2388 and the oncoprotein p185HER-2 in patients with systemi-
Marino P, Preatoni A, Cantoni A (1995) Randomized trials of cally untreated non-small-cell lung cancer: an immunohis-
radiotherapy alone versus combined chemotherapy and tochemical study on cryosections. Br J Cancer 74:86–91
radiotherapy in stages IIIa and IIIb non-small cell lung Pollmann M, Parwaresch R, Adam-Klages S et al (2006) Human
cancer. A meta-analysis. Cancer 76:593–601 EML4, a novel member of the EMAP family, is essential for
Mascaux C, Iannino N, Martin B et al (2005) The role of RAS microtubule formation. Exp Cell Res 312:3241–3251
oncogene in survival of patients with lung cancer: a Potti A, Mukherjee S, Petersen R et al (2006) A genomic
systematic review of the literature with meta-analysis. Br strategy to refine prognosis in early-stage non-small-cell
J Cancer 92:131–139 lung cancer. N Engl J Med 355:570–580
Mattson K, Holsti LR, Holsti P et al (1988) Inoperable non- Potti A, Mukherjee S, Petersen R et al (2011) Retraction: a
small cell lung cancer: radiation with or without chemo- genomic strategy to refine prognosis in early-stage non-
therapy. Eur J Cancer Clin Oncol 24:477–482 small-cell lung cancer. N Engl J Med 364:1176
Mayne ST, Buenconsejo J, Janerich DT et al (1999) Familial Ramalingham SS, Owonikoko TK, Khuri FR (2011) Lung
cancer history and lung cancer risk in USA non smoking cancer: new biological insights and recent therapeutic
men and women. Cancer Epidemiol Biomarkers Prev advances. Ca Cancer J Clin 61:91–112
8:1065 Reed E (1998) Platinum–DNA adduct, nucleotide excision
Meyerson M, Carbone D (2005) Genomic and proteomic repair and platinum based anti-cancer chemotherapy.
profiling of lung cancers: lung cancer classification in the Cancer Treat Rev 24:331–344
age of targeted therapy. J Clin Oncol 23:3219–3226 Riely GJ, Marks J, Pao W (2009) KRAS mutations in non-small
Mitsudomi T, Hamajima N, Ogawa M et al (2000) Prognostic cell lung cancer. Proc Am Thorac Soc 6:201–205
significance of p53 alterations in patients with non-small Rodenhuis S, van de Wetering ML, Mooi WJ et al (1987)
cell lung cancer: a meta-analysis. Clin Cancer Res 6:4055– Mutational activation of the K-ras oncogene. A possible
4063 pathogenetic factor in adenocarcinoma of the lung. N Engl J
Mitsudomi T, Morita S, Yatabe Y et al (2010) Gefitinib versus Med 317:929–935
cisplatin plus docetaxel in patients with non-small-cell lung Rosell R, Danenberg KD, Alberola V et al (2004) Ribonucle-
cancer harbouring mutations of the epidermal growth factor otide reductase messenger RNA expression and survival in
receptor (WJTOG3405): an open label, randomised phase 3 gemcitabine/cisplatin-treated advanced non-small cell lung
trial. Lancet Oncol 11:121–128 cancer patients. Clin Cancer Res 10:1318–1325
Mitsudomi T, Steinberg SM, Oie HK et al (1991) Ras gene Rosell R, Moran T, Queralt C et al (2009) Screening for
mutations in non-small cell lung cancers are associated with epidermal growth factor receptor mutations in lung cancer.
shortened survival irrespective of treatment intent. Cancer N Engl J Med 361:958–967
Res 51:4999–5002 Rusch V, Klimstra D, Venkatraman E et al (1997) Overex-
Miyake Y, Kodama T, Yamaguchi K (1994) Pro-gastrin- pression of the epidermal growth factor receptor and its
releasing peptide (31–98) is a specific tumor marker in ligand transforming growth factor alpha is frequent in
patients with small cell lung carcinoma. Cancer Res resectable non-small cell lung cancer but does not predict
54:2136–2140 tumor progression. Clin Cancer Res 3:515–522
808 D. Singh et al.

Sasaki T, Rodig SJ, Chirieac LR et al (2010) The biology and Taguchi F, Solomon B, Gregorc V et al (2007) Mass
treatment of EML4-ALK non-small cell lung cancer. Eur J spectrometry to classify non-small-cell lung cancer patients
Cancer 46:1773–1780 for clinical outcome after treatment with epidermal growth
Sause W, Kolesar P, Taylor S IV et al (2000) Final results of factor receptor tyrosine kinase inhibitors: a multicohort
phase III trial in regionally advanced unresectable non- cross-institutional study. J Natl Cancer Inst 99:838–846
small cell lung cancer. Chest 117:358–364 Tapia FJ, Polak JM, Barbosa AJ et al (1981) Neuron-specific
Scartozzi M, Franciosi V, Campanini N et al (2006) Mismatch enolase is produced by neuroendocrine tumors. Lancet
repair system (MMR) status correlates with response and 1:808–811
survival in non-small cell lung cancer (NSCLC) patients. Thatcher N, Chang A, Parikh P et al (2005) Gefitinib plus best
Lung Cancer 53:103–109 supportive care in previously treated patients with refractory
Schaake-Koning C, van den Bogaert W, Dalesio O et al (1992) advanced non-small cell lung cancer: results from a
Effects of concomitant cisplatin and radiotherapy on randomized placebo controlled multicenter study (Iressa
inoperable non-small cell lung cancer. N Eng J Med 326: survival evaluation in lung cancer). Lancet 366:1527–1537
524–530 Volm M, Efferth T, Mattern J (1992) Oncoprotein (c-myc,
Schiller JH, Adak S, Feins RH et al (2001) Lack of prognostic c-erbB1, c-erbB2, c-fos) and suppressor gene product (p53)
significance of p53 and K-RAS mutations in primary expression in squamous cell carcinomas of the lung.
resected non- small-cell lung cancer on e4592: a laboratory Clinical and biological correlations. Anticancer Res
ancillary study on an eastern cooperative oncology group 12:11–20
prospective randomized trial of postoperative adjuvant Westfall JM, Mold J, Fagnan L (2007) Practice-based research-
therapy. J Clin Oncol 19:448–457 ‘‘Blue Highways’’ on the NIH roadmap. JAMA 297:403–
Shaw AT, Yeap BY, Mino-Kenudson M et al (2009) Clinical 406
features and outcome of patients with non-small-cell lung Woenckhaus M, Stoehr R, Dietmaier W et al (2003) Micro-
cancer who harbor EML4-ALK. J Clin Oncol 27:4247–4253 satellite instability at chromosome 8p in non-small cell lung
Shih CM, Chen CY, Lee IH et al (2010) A polymorphism in the cancer is associated with lymph node metastasis and
hMLH1 gene (-93G [ A) associated with lung cancer squamous differentiation. Int J Oncol 23:1357–1363
susceptibility and prognosis. Int J Mol Med 25:165–170 Wójcik E, Kulpa JK, Sas-Korczyńska B (2008) ProGRP and
Simon G, Sharma A, Li X et al (2008) Feasibility and efficacy NSE in therapy monitoring in patients with small cell lung
of molecular analysis-directed individualized therapy in cancer. Anticancer Res 28:3027–3033
advanced non-small-cell lung cancer. J Clin Oncol 25: Wu Q, Christensen LA, Legerski RJ et al (2005) Mismatch
2741–2746 repair participates in error-free processing of DNA inter-
Sinclair WK, Morton RA (1966) X-ray sensitivity during the strand crosslinks in human cells. EMBO Rep 6:551–557
cell generation cycle of cultured Chinese hamster cells. Xinarianos G, Liloglou T, Prime W et al (2000) hMLH1 and
Radiat Res 29:450–474 hMSH2 expression correlates with allelic imbalance on
Skarda J, Fridman E, Pevova P et al (2006) Prognostic value of chromosome 3p in non-small cell lung carcinomas. Cancer
hMLH1 and hMSH2 immunohistochemical expression in Res 60:4216–4221
non-small cell lung cancer. A tissue microarray study. Yamaguchi K, Abe K, Kameya T et al (1983) Production and
Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub molecular size heterogeneity of immunoreactive gastrin-
150:255–259 releasing peptide in fetal and adult lungs and primary lung
Slebos RJ, Kibbelaar RE, Dalesio O et al (1990) K-ras tumors. Cancer Res 43:3932–3939
oncogene activation as a prognostic marker in adenocarci- Yang LY, Li L, Jiang H et al (2000) Expression of ERCC1
noma of the lung. N Engl J Med 323:561–565 antisense RNA abrogates gemcitabine-mediated cytotoxic
Souglakos J, Boukovinas I, Taron M et al (2008) Ribonucle- synergism with cisplatin in human colon tumor cells
otide reductase subunits M1 and M2 mRNA expression defective in mismatch repair but proficient in nucleotide
levels and clinical outcome of lung adenocarcinoma patients excision repair. Clin Cancer Res 6:773–781
treated with docetaxel/gemcitabine. Br J Cancer 98: Yarden Y, Sliwkowski MX (2001) Untangling the ErbB
1710–1715 signalling network. Nature Rev Mol Cell Biol 2:127–137
Steels E, Paesmans M, Berghmans T et al (2001) Role of p53 as Yin Y, Tainsky MA, Bischoff FZ et al (1992) Wild-type p53
a prognostic factor for survival in lung cancer: a systematic restores cell cycle control and inhibits gene amplification in
review of the literature with a meta-analysis. Eur Respir J cells with mutant p53 alleles. Cell 70:937–948
18:705–719 Zhang H, Hyrien O, Pandya K et al (2008) Tumor response
Sugio K, Ishida T, Yokoyama H et al (1992) Ras gene kinetics after schedule-dependent paclitaxel chemoradiation
mutations as a prognostic marker in adenocarcinoma of the treatment for inoperable non-small cell lung cancer: a model
human lung without lymph node metastasis. Cancer Res for low-dose chemotherapy radiosensitization. J Thorac
52:2903–2906 Oncol 3:563–568
Sun Z, Wigle DA, Yang P (2008) Non-overlapping and non- Zheng Z, Chen T, Li X et al (2007) DNA synthesis and repair
cell-type-specific gene expression signatures predict lung genes RRM1 and ERCC1 in lung cancer. N Engl J Med
cancer survival. J Clin Oncol 26:877–883 356:800–808
Clinical Research in Radiation Oncology
of Lung Cancer: Why We Fail(ed)?
Branislav Jeremić

Contents Abstract
Clinical research in radiation oncology of lung
1 Introduction.............................................................. 809 cancer can bring important advances in the field of
2 Clinical Research Generals .................................... 810 optimized treatment approaches in this disease.
However, such a research has been a subject of many
3 Technological Addictions ........................................ 810
controversies, in particular in the light of lack of
4 Clinical Failures....................................................... 812 adequate number and quality of clinical trials which
5 Controversial Issues ................................................ 813 could have changed our standard policies. To
6 Possible Solutions..................................................... 817
address the comprehensive issue of clinical research
in radiation oncology of lung cancer, several aspects
References.......................................................................... 817
will be considered: clinical research generals,
technological addictions, clinical failures, and some
of existing, rather controversial issues identified.
Finally, some of possible solutions for the current,
grossly unfavorable, situation will be discussed.

1 Introduction

Lung cancer remains one of the most important health


problems worldwide. This is not so, only because of its
high morbidity and mortality (as the major cancer
killer in both sexes), but also due to the fact that dismal
figures obtained nowadays reflect largely inadequate
efforts to optimize our prevention, screening, early
diagnosis, treatment, and palliation efforts in this dis-
ease. Radiation therapy remains one of the corner-
stones of modern therapeutic endeavours due to its
high effectiveness as both curative and palliative
approach given either alone or in combination with
other therapeutic modalities (surgery, chemotherapy)
in the majority of patients with this disease.
B. Jeremić (&)
Institute of Lung Diseases, Sremska Kamenica, Serbia However, our ability to theoretically consider all
e-mail: nebareje@gmail.com possible avenues to optimize radiation therapy and then

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 809
DOI: 10.1007/174_2011_222, Ó Springer-Verlag Berlin Heidelberg 2011
810 B. Jeremić

practically apply it in daily clinic has been very low. combined radiochemotherapy in locally advanced
Hence, improvements have been only incremental and nonsmall cell lung cancer, only a few prospective
suffice to say, inadequate, having in mind that each year phase III clinical trials have been successfully per-
more than one million patients are newly diagnosed with formed, fully analyzed and fully published (Belderbos
this disease. The vast majority of lung cancer patients do et al. 2007; Curran et al. 2000; Sause et al. 1995;
not benefit from any of these ‘‘improvements’’ irre- Schaake-Koning et al. 1992). As probably the most
spective of geography, race or social status/income. extreme example is the case of extensive disease
Therefore, lung cancer must be considered as one of the small cell lung cancer where we have had a single
major battlegrounds in clinical research of oncology, prospective randomized trial ever reporting on the
since clinical research can bring important discoveries role of curative thoracic radiation therapy in this
and help improve our ability to more successfully eval- disease. As an additional evidence supporting these
uate more general oncological principles in this disease. statements is the fact that after publication of (Jeremic
This also relates to continuous and very much evolving et al. 1999) on radiation therapy in extensive disease
evaluation of the place and the role of radiation therapy small cell lung cancer, 10 years passed before two
in the field of clinical research in thoracic oncology. studies opened up for patient accrual in the US and
To address the comprehensive issue of clinical The Netherlands, respectively. Finally, when one
research in radiation oncology of lung cancer, several remains in the same time period (e.g. last three decade),
aspects will be considered: clinical research generals, it must be clearly observed that what and when have
technological addictions, clinical failures, and some driven researchers is less the idea itself, but, unfortu-
of existing, rather controversial issues identified. nately, more fashion and trend. Hence, we have seen an
Finally, some of possible solutions for the current, avalanche of induction chemotherapy studies in the
grossly unfavorable, situation will be discussed. 1980s of the last century (Dillman et al. 1990; Le
Chevalier et al. 1992; Clamon et al. 1999; Vokes et al.
2002; Belani et al. 2005a) and many of the consolida-
2 Clinical Research Generals tion chemotherapy studies in the 1990s of the last
century (Lau et al. 2001; Gandara et al. 2006; Sekine
General observations can be easily understood if one et al. 2006; Hanna et al. 2008), all asking very similar
tries to answer the following questions: who does questions, with very similar design, majority of which
clinical research, how one does it and finally, when were not asking strategically different question from
one does it? Putting events (e.g. clinical trials their predecessors. Changing a regimen consisting of
designed and performed) into a chronological order, 70 mg/sqm of paclitaxel given every week with
instant observation is that the vast majority of clinical 100 mg/sqm of cisplatin given every 3 weeks to a
research in radiation oncology of lung cancer (and regimen consisting of 60 mg/sqm of paclitaxel given
very much with other treatment options) through once a week with 50 mg/sqm of cisplatin given twice a
clinical trials is mostly done within single-institu- week meant very little, if anything. Indeed, it mostly
tional setting and less within the group setting and brought nothing to the overall scientific improvements
even less within the intergroup setting. This, unfor- in this field, not to mention that thousands of patients
tunately, leads to less patients enrolled per study and have been exposed to meaningless clinical research that
that, ultimately, leads to poorer (lower) clinical evi- ‘‘misused’’ millions of dollars and Euros worldwide.
dence such studies can bring. Therefore, it must be
clearly said that this type of approach leads to inferior
quality of clinical research and hampers implemen- 3 Technological Addictions
tation into a general clinical practice of radiation
oncology of lung cancer. Similarly, when answering Radiation oncology is technology driven and tech-
to a question how one does it, again, more phase I and nology dependent profession. It was so since the
II trials are observed and less phase III trials are advent of X-rays and it will definitely remain so in the
performed. For example, if one takes as an example of foreseeable future. The level of this dependency is
what two of the major groups (RTOG and EORTC) very much related to several factors. Some of them
have done in the last 30 years in the field of e.g. are inherent to national health care systems (due to
Clinical Research in Radiation Oncology of Lung Cancer: Why We Fail(ed)? 811

billing specifics), while some are related to the pro- industry in the field of clinical research of radiation
fessional and/or scientific level of an institution. oncology in lung cancer, sometimes this influence is
While free-standing/private institutions usually see at least hard to understand. While marketing aspects
technological advances as a driving force for more bring necessary flavour to the overall happenings in
patients expecting that patient flow remain crucially the field, sometimes they do the harm. I will here
dependent on offering the latest available technology briefly describe the situation occurring during one of
(and assuming that these technologies work well the major European meetings which occurred several
against cancer), academic institutions are usually years ago. During evening session organized as a pure
inclined towards newer technologies also due to marketing event for hundreds of guests, one machine
enormous capacity for research such technologies company marketed their product using the slogan:
usually bring early in the phase of the development. ‘‘Problem solving for tumor motion’’. What made this
In both cases mindset towards ‘‘new’’ (presumably slogan a bit inaccurate was not the machine itself, or
also being better!) is what actually drives clinicians its future and potential capabilities (to be clinically
and scientists and that is the fact worldwide. Let us proven) but rather supporting the slogan by identify-
briefly take the matter of introduction and subsequent ing a person saying so, an unusual matter in a sci-
use of intensity modulated radiation therapy in the entific world, at least in such marketing aspect.
clinical practice worldwide, especially in the US. Due Finally, what made this approach really problematic
to the fact that it had found fast way to overall and was that the person (radiation oncologist) whose
legal approval, with very favorable billing profile, this name was used to support this marketing effort never
technology became so widely available that majority did anything serious in the field of lung cancer and
of institutions started almost indiscriminately using it was actually known as head and neck person due to
in majority of tumor sites. Logical question (evidence good achievements in the field of head and neck
vs. type of the proof needed) one may ask is whether we radiation oncology. It was obvious that company
actually have had ‘‘evidence’’ to support this? Without embarking on this task had nobody from the field of
going into a detail what ‘‘evidence’’ actually means, we lung cancer able to say those words, likely because
faced the fact that only a few clinical trials have actually real professional in the field would not uncondition-
compared in a prospective randomized fashion this ally support unproven technologies, no matter how
technology with already available technologies such as promising they look. Contrary to this technology,
2D or 3D radiation therapy. In spite of being rather another one was popularized much less over the
complex and time consuming application (evidence vs. years. In spite of several vendors being active in the
challenge), clinicians nevertheless continued with its field, offering different products, various stereotactic
use in clinic, largely based on the fact that ‘‘isodose applications proved to be extremely and almost
distribution’’ (or DVH or else) achieved with it was identically efficient in the treatment of both primary
better than with other technologies’’. Again, one can (early) and metastatic nonsmall cell lung cancer. This
rightly point out that what we actually do is nothing but became obvious when one looks at how different dose
using a surrogate (in this case computer screen repre- prescriptions, fractionation patterns and immobiliza-
sentation of physical calculations done on phantoms) of tion devices have been used worldwide, all leading to
the real life (clinic, better to say) as definite measure of very similar results.
comparison between the two. So, what is done this way Finally, technological addictions will also have an
it is nothing but the third in a row of hurdles, one may inadvertent influence on radiobiological aspects of
call evidence versus intention. Whichever evidence we contemporary clinical fractionation. With more ori-
confront with any comparator, the ultimate question, in entation towards larger dose per fraction and, hence,
every single tumor site, would simply remain whether shorter regimens, these hypofractionated regimens
there are Clinically Meaningful benefits for this and would prove as unexplored territory for combined
other more advanced cases to justify its discriminate radiochemotherapy and I particularly refer to con-
use in daily clinic. current radiation therapy and chemotherapy. What we
One may also point out the active influence of mostly practice today is based on our experience
industry in this regard. While this author fully gathered when ‘‘classic’’ radiobiology modelling and
understands and supports active engagement of clinical studies were combined to gather knowledge
812 B. Jeremić

that serves as the cornerstone of the current evidence- why we do not have more patients enrolled into
based decision–making process. That said, standard clinical trials since the vast majority of clinical trials
dose fraction radiation therapy and chemotherapy as are continuously used to ask various questions about
we have used it and still use, may mean little in the optimization via intensification of radiation therapy
future when large dose per fraction and fewer times and/or chemotherapy.
available (over the course of combined radiation Of tumor-related factors, stage, histology, and
therapy and chemotherapy) to administer chemo- location can be used to provide necessary framework
therapy concurrently with radiation therapy. While for statements above. Stage III nonsmall cell lung
this may lead to fewer concurrent regimens in the cancer is one of the focuses of clinical investigations
future, it may also lead to somewhat higher incidence in the field of lung cancer. Even with the most recent
of both acute and late toxicity such ‘‘hybrid’’ regi- staging revision (Goldstraw et al. 2007), it still has 11
mens may lead to due to larger dose per fractions and different T and N combinations. While we continue
less time to fractionated total chemotherapy dose. this, rather surgical, staging system, it is unknown
why radiation oncologists do not embark on first
evaluating and then using more tumor volume-, rather
4 Clinical Failures than tumor size-driven staging system(s). This is
especially so since we are living for, at least, the last
The term ‘‘clinical failures’’ is used here to describe 20 years, in an era of powerful computers which
our failures to observe events during clinical studies enabled easy 3D reconstruction of various volumes
performed in the past and to use then-gathered and, hence, fast computation of volumetrics. While
knowledge to optimize subsequent studies. In partic- occasional work had been done mostly concentrating
ular, this is referred to the time component to use on T volumes, N volumes seem as almost completely
gathered knowledge to optimize future attempts via neglected matter. Regarding histology, it has been
clinical studies. These failures can broadly be sepa- speculated for many years about more ‘‘local’’ char-
rated into patient-related, tumor-related, and history acter of the squamous cell carcinomas than other
of the disease-related. nonsmall cell carcinomas (e.g. adenocarcinoma, large
Of patient-related observations, it was frequently cell carcinoma). If this is so, why then efforts have not
observed that patients with performance status of less been appropriately made towards optimization of
than 50% and/or those with pronounced weight loss our endeavours by using more local (e.g. radiation
(i.e. [5%) are those suffering from more toxicity, therapy dose escalation with/without radioenhancing
having more treatment interruptions and/or dose administration of chemotherapy) treatment options in
reductions, all leading to poorer local control and this setting and, contrary to that, more/higher doses of
ultimately, inferior survival. Hence, it may come as a chemotherapy given with radiation therapy in ade-
surprise that although more recent studies tried as nocarcinomas and large cell carcinomas due to their
much as possible to exclude these patients from var- presumably larger metastatic potential? Similarly,
ious dose-intensification trials, we do not have more some observed more ‘‘local’’ character of peripheral
clinical studies concentrating on those, rather unfa- lung cancers, especially early stage ones. Therefore,
vorable, prognostic group with only a few exemptions different approaches could have been explored in
to this statement. Similarly, elderly have largely been addressing these issues and helped gain insight about
neglected adequate access to clinical trials, possibly differences in both biological behavior of tumors
due to a fear that they could not tolerate intensive located differently as well as design clinical trials
therapeutic interventions. While there are no firm data addressing different therapeutic options tailored
supporting this, rather nihilistic view, there are also according to these characteristics.
not many prospective randomized clinical trials spe- Of those observations that could have easier
cifically addressing various aspects of treatments changed our reality by using them in clinic, events in
administered in the field of radiation oncology of lung locally advanced nonsmall cell lung cancer and
cancer, where radiation therapy was given either extensive disease small cell lung cancer could nicely
alone or in combination with chemotherapy. These serve as supporting background. In locally advanced
observations may fall into a group of explanations of nonsmall cell lung cancer, induction chemotherapy
Clinical Research in Radiation Oncology of Lung Cancer: Why We Fail(ed)? 813

followed by radical radiation therapy became stan- over induction chemotherapy and radiation therapy,
dard treatment option in the 1990s of the last century. there are still clinicians embarking on this hazardous
In order to optimize it, various intensification and inferior treatment option, driven, obviously, by
attempts have been made, including administration of other than evidence-based motives. Similarly, by
concurrent radiochemotherapy after induction che- observing patterns of failure in extensive disease
motherapy. What all of these studies clearly identified small cell lung cancer treated with chemotherapy
is insufficient local control obtained when one starts alone, (Jeremic et al. 1999) designed and executed a
the treatment with induction chemotherapy, no matter prospective clinical trial that successfully integrated
how much you eventually intensify the second (con- radical chest radiation therapy in combined modality
current) part of the combined treatment with either approach. While this study was praised as pioneering
higher radiation therapy dose or more drugs/doses, one, researchers continued to practice chemotherapy
some of which belong to the third generation drugs. alone in this disease. It took about 10 years after full
What was occasionally discussed in the literature publication of the date of (Jeremic et al. 1999) to
became obvious with the study of (El Sharouni et al. embark on similar studies evaluating the role of
2003) who compared CT scans pre-and post-induction radiation therapy in this disease.
chemotherapy, with emphasis on the time from the
last induction chemotherapy cycle to the time of
radiation therapy treatment planning CT was actually 5 Controversial Issues
done. That way they have been able to measure an
increase in gross tumor volumes (GTV) and subse- They can broadly be divided into pressure from
quently define volume doubling times. During the industry (largely discussed above), hospital/depart-
waiting period (for the planning CT scan and start of mental workflow, and some personal issues. Hospital/
RT), a total of 41% of all tumors became incurable, departmental workflow was and still seems to be one
with the ratio of GTVs being in the range of 1.1–81.8! of the major problems in clinical research in the field
Tumor doubling times ranged 8.3–171 days, with the of lung cancer worldwide. They include sometimes
median of 29 days. When translated into more clini- existing waiting lists which adversely influence time
cal language, these findings clearly say that even if to start with radiation therapy treatments and, before
one may have thought (due to insufficient CT-based it, treatment planning activities. In the past, this was
imaging) that response occurs (and that it matters), frequently discriminatory issue, which was mostly
there is actually an opposite development, with sur- used by medical oncologists and/or pneumologists as
viving tumor clonogens repopulating fast, leading the driving force to start combined treatment with
tumors to regrow to the state of incurability. When administering chemotherapy first. While the nature of
applied to observations of various clinical studies, the work of medical oncologists/pulmonologists
whatever you do after you start with chemotherapy, it dealing with lung cancer remains the domain of
became widely known that failure is inevitable and chemotherapy from the onset, with a paradigm, ‘‘the
comes fast. With this approach, one can only achieve more of it, the better of it’’, radiation oncologists
more toxicity (Vokes et al. 2002; Akerley et al. 2005; have, unintentionally, I assume, added to these fail-
Socinski et al. 2008) and even if you use the modern ures by accepting the policy that we ‘‘should do
radiation therapy tools such as 3D radiation therapy something’’ while patient is waiting for planning CT
and attempt treatment intensification by escalating the scan, as part of the preparation before the radiation
total dose, again, one cannot achieve better outcomes. therapy will start. We have indeed done/achieved
Indeed, impressive 12% mortality in the most recent something! Several clinical trials clearly showed that
CALGB attempt (Socinski et al. 2008) to combine any intensification of the latter/major part of the
induction chemotherapy with subsequent radiation combined treatment approach via concurrent radio-
therapy and concurrent chemotherapy led investiga- chemotherapy is not effective, once you have started
tors to early stopping the trial. Interesting, though, is with induction chemotherapy. Several studies of
that even nowadays, after successful publication of CALGB (Vokes et al. 2002; Akerley et al. 2005;
three meta analyses which clearly showed superiority Socinski et al. 2008), showed that there is no com-
of concurrent radiation therapy and chemotherapy pensation for insufficient start (with chemotherapy).
814 B. Jeremić

Other attempts, such as the use of three daily fractions pneumologist. While this presents no problem for
of radiation therapy, also proved to be ineffective. All doubtful cases, those with definite diagnosis needed,
in all, whatever you do after you start with chemo- or those of borderline cases, it remains absolutely
therapy, failure is inevitable and comes fast. With this unknown why fully staged patient with stage III
approach, you can only achieve more toxicity such as nonsmall cell lung cancer, having a performance
12% mortality in the most recent CALGB (30105) status score of e.g. 80%, no weight loss, squamous
(Socinski et al. 2008) attempt to combine induction histology and no pulmonary reserve compromised
chemotherapy with subsequent radiochemotherapy (provided that all of these had been previously
that led investigators to early stopping the trial. investigated and confirmed) is not sent to radiation
Waiting lists remain to be an unpleasant reality in oncologist who would then be responsible for the
radiation oncology departments in various regions all overall treatment plan including chemotherapy. This
over the world. While one can instantly pinpoint at is even more so in inoperable cases with serious lung/
them as one of the sources of potential obstacle for heart issues when neither surgery nor chemotherapy
implementing e.g. concurrent radiation therapy and would be offered. Including another specialist leads
chemotherapy, there are measures to overcome it, only to loosing additional time, as well as it will
either completely, or at least significantly. This is obscure clear picture and undermine efforts of one
especially so if one continues to use long and pro- efficient treatment option, namely radiation therapy.
tracted regimens in the treatment of metastatic disease Perhaps, radiation oncologists should go more to
or even locally advanced incurable cases. Therefore, public, inform it about standards of care, engage in
there is little justification and sympathy for those continuous discussions with other potential sources of
radiation oncologists which continue to use e.g. patient referrals (e.g. general physicians, respiratory
30 Gy in 10 fractions for a single painful bone physicians working outside academic/university hos-
metastasis (instead of using 8 Gy in a single fraction) pital/centres) and bring more awareness to non-radi-
and, at the same time, complain about existing wait- ation oncology community about opportunities of
ing lists. To extend this, some of brain metastasis modern radiation oncology in the field of lung cancer.
patients could effectively be treated with 20 Gy in Some of the controversial issues are actually per-
five fractions and some of locally advanced incurable sonal in nature. We all occasionally witness evidence-
lung cancer with either 10 Gy in a single fraction or based versus expert-based opinions. Unfortunately,
17 Gy in two fractions given with 1 week split. While even some of the most prestigious institutions all over
billing issues still dominate as one of the reason for the world suffer from this disease including also
favoring more protracted regimens, hospital/depart- inherent specifics of medical profession and division
ment management should prioritize access to radia- between ‘‘big’’ specialities (e.g. surgery, internal
tion therapy-based on cost-effectiveness, at least. medicine) and presumably ‘‘smaller’’ (e.g. radiation
In addition, worldwide experience with routes of oncology). I am sure all of thoracic radiation oncol-
patient referral for lung cancer treatment remains ogists occasionally witnessed unpleasant situations in
unfavorable for radiation oncologist. In spite of the daily clinic or during tumor board meetings where
fact that radiation oncologists actively working in the this has occurred. Collateral damage may well be our
field of stereotactic radiation therapy have seen more residents, who are primed with this type of really
referrals directly to them, largely due to excellent unacceptable behavior for which there should be only
results achieved with this treatment technique, this is a zero tolerance. Extension to this is its soft version
not the case when more standard (here meant as three- materialized in frequent existence (among radiation
dimensional conformal radiation therapy or intensity oncologists) of ‘‘non-believers’’ in evidence-based
modulated radiation therapy) approach in both early principles. Some people simply do not believe evi-
and locally advanced nonsmall cell lung cancer and dence they are presented with. Usually, this is not
most of limited disease small cell lung cancer patients related to ‘‘constructive’’ (better said, productive)
is considered. There, besides official and visible criticism, based on scientific merits, but rather finding
tumor boards, there seems to exist an invisible one, excuses of how to interpret existing evidence. Even
materialized in the fact that patients are first referred softer version of non-believing in existing evidence is
to either thoracic surgeon and/or medical oncologist/ seen in some of our colleagues who request always
Clinical Research in Radiation Oncology of Lung Cancer: Why We Fail(ed)? 815

more evidence than is present and/or needed. Excuses and large non-academic institutions/practicing physi-
goes from: (1) ‘‘this is only a second trial confirming cians would feel some heat for non-recruiting patients
that A is better than B’’, through (2) ‘‘logistics of our into the juicy clinical studies investigating novel
clinic prevent us of implementing this approach (e.g. drugs or combinations. Another matter is how fast this
bid fractionation)’’ to (3) ‘‘the statistical power of that can be observed in daily routine. It is hard completely
particular study is not strong enough’’. Even in cases to digest authors explanation that e.g. LAMP study
of existing meta-analyses and multiple large confir- (Belani et al. 2005b) widely influenced practice in the
matory trials there may be an obstacle to implement US being published only as an abstract in the year
what is already proven. Several years ago, (Langer 2002 (and had, therefore, only several months of its
et al. 2005) report on the chemotherapy characteris- limited abstract life to exert influence on troublesome
tics practiced in nonmetastatic lung cancer in the U.S. behavior of practicing physicians in this Patterns of
during the Patterns of Care Survey survey in 59 Care Study, published in 2004) while, on the other
radiation therapy institutions. Informations have been side, RTOG 9410 (Curran et al. 2000) did not exert
sampled during a period 2000–2002. Only 6% of such an influence, being published in the same way
patients received hyperfractionated (b.i.d.) radiation (abstract) in the year 2000! Speed of such an imple-
therapy in small cell lung cancer, only 22% received mentation is more than debatable.
prophylactic cranial irradiation and only 49% patients Sometimes, however, evidence is confusing, even
underwent CT-based treatment planning. Signifi- when coming from three meta-analyses as we have
cantly more patients older than 70 years were treated recently seen when focusing on the issue of timing of
at large nonacademic facilities compared with smaller administration of radiation therapy and chemotherapy
nonacademic or academic institutions, surprising in limited-disease small cell lung cancer. While one
finding for non-treating institutions since a large body may assume that all three analyses showed the same
of data exists on the effectiveness of radiation therapy outcome, based on the same data from the available
in elderly or simply a finding underlining the lack of literature, however, they do not show that. If one
radiation therapy studies in this patient population. roughly compares their conclusions, the meta analysis
Some 5% patients with small cell lung cancer do not by (Huncharek and McGarry 2004) showed the
receive chemotherapy, while 18% of stage I nonsmall highest differential survival advantage for early ver-
cell lung cancer patients receive radiation therapy/ sus late radiation therapy and chemotherapy, a sys-
chemotherapy and this percentage goes up to 34% for tematic review by (Fried et al. 2004) showed a
stage II nonsmall cell lung cancer. Of additional somewhat smaller but nevertheless significant
importance is that 33% of locally advanced nonsmall advantage for early radiation therapy and chemo-
cell lung cancer receives only radiation therapy. therapy (greater at 2 years than at 3 years), while a
Induction chemotherapy is still widely practiced, systematic review by (Pijls-Johannesma et al. 2004)
while trend seems to have favored consolidation concluded that ‘‘the optimal integration of radiation
chemotherapy as well. These examples are perfect therapy and chemotherapy in limited disease small
show of practice of non-evidence-based medicine in cell lung cancer is unknown’’. How does one recon-
lung cancer as they also nicely match observation cile these findings? Is it simply 2 outweighing 1? Not
from the choice of timing of radiation therapy/che- necessarily. There are a number of facts which need to
motherapy in small cell lung cancer, choice of drugs, put into the right perspective, especially in the review
etc. They all fortify general impression that although of (Pijls-Johannesma et al. 2004) which give addi-
combined radiation therapy and chemotherapy are tional insight into this matter. It is, therefore, that,
practiced by the majority of institutions in majority of sometimes ‘‘meta-analysis of the meta-analyses’’,
locally advanced nonsmall cell lung cancer and small may be needed, although this could rightly be criti-
cell lung cancer patients, there are still tremendous cised as additional example of expert-based medicine!
variations in practice, most of which are not sub- (Jeremic B. 2006). Basically, there are various dif-
stantiated by the existing evidence. It remained ferences in these three analyses. Some of them are
unknown whether perhaps group affiliation of the inherent to the characteristics of the accepted studies,
sampled radiation therapy facilities influenced the such as a different definition of the ‘‘limited disease
practice, because it is likely to expect that academic small cell lung’’ which included more or less of the
816 B. Jeremić

‘‘distant’’ intrathoracic nodal areas, as well as differ- is that even those that are with clear outcomes and
ent radiation therapy and chemotherapy characteris- great potential for improving overall approach in lung
tics. Others are inherent in the analyses themselves, cancer are frequently poorly implemented in practice.
such as a different definition of ‘‘early’’ and ‘‘late’’ One good example is aforementioned hyperfraction-
radiation therapy and chemotherapy. These led to ation trial in limited disease small cell lung cancer.
different inclusion and exclusion criteria which Another good example may well be the so-called
resulted in different numbers of patients being inclu- CHART (continuous hyperfractionated accelerated
ded into these analyses, enhancing great heterogeneity radiation therapy) which produced significant
among the studies, a usual finding when a meta- improvement in treatment outcome when compared to
analysis is done. Also, although all used meta-analytic standard radiation therapy in inoperable nonsmall cell
approach, there were differences in statistical inter- lung cancer (Saunders et al. 1999). Due to its poor
ventions and subgroup analyses, and outcomes used implementation, even in the UK, the National Health
(local control, toxicity). Finally, none of the three Service (NHS) there had to officially urge institutions
analyses used individual patient data. Contrary to to adopt such an approach. Also, as we mentioned
the two analyses (Huncharek and McGarry 2004; above, some of the trials implemented in clinical
Fried et al. 2004) observing significant advantage practice are not those representing standards of
for ‘‘early’’ radiation therapy and chemotherapy over treatment as various Patterns of Care Studies have
the ‘‘late’’ radiation therapy and chemotherapy, the shown (Movsas et al. 2003; Langer et al. 2005).
review of (Pijls-Johannesma et al. 2004) was not Finally, an intriguing thing for clinicians and
supportive of these findings. Since that review (2004) researchers in this field is frequent confusion about
indicated no difference between the ‘‘early’’ and the the ways to optimize existing standards. For example,
‘‘late’’ schedules, one must turn to other outcome after Intergroup study (Turrisi et al. 1999) clearly
measures to have better perspective about therapeutic established hyperfractionation as the superior
benefit of these two approaches. Interestingly, toxicity approach in limited disease small cell lung cancer,
data showed that there was no difference between investigators have not embarked on further optimi-
outcomes for ‘‘early’’ and ‘‘late’’ schedules regarding zation of this, superior, approach, but rather on opti-
various types of toxicity, except that leucopoenia was mization of inferior (currently once-daily radiation
significantly higher in the ‘‘late’’ group. This finding therapy) through studies of RTOG/CALGB and EO-
would favor ‘‘early’’ administration of radiation RTC, respectively. This way, no matter how outcome
therapy and chemotherapy having better overall out- of the two ongoing studies may look like in the next
come than ‘‘late’’ radiation therapy and chemother- 3–5 years, we have already lost precious time in: (1)
apy. Finally, a subgroup analysis might be done to poorly practicing hyperfractionation in daily clinic
focus on an important issue (e.g. fractionation or type although it is de facto standard of treatment, (2)
of chemotherapy) or when a seemingly inappropriate designing trials that may perhaps (e.g. after some
study (due to different reasons) was excluded from an 15 years) show superiority of high-dose standard
analysis, the improved homogeneity of the remaining fraction radiation therapy over Intergroup hyperfrac-
studies instantly led to a favorable effect of ‘‘early’’ tionation schedule, and (3) lead us again to the issue
radiochemotherapy. It is obvious that more ‘‘consol- of more clinical research within the same loop,
idation’’ efforts should be directed towards bringing e.g. hyperfractionation, to be tested in the next
the highest level of evidence to the community of 10–15 years (from now) with e.g. 54 Gy in 36 frac-
thoracic oncologists and that we should, perhaps, pay tions in 18 treatment days and concurrent low-dose
additional effort and add additional forces to sys- chemotherapy, as this author successfully imple-
tematically address issues of importance for our mented in both limited- and extensive-disease small
profession. cell lung cancer (Jeremic et al. 1997, 1999). Suffice to
A number of controversial issues could also say, with this approach, we may end up in the e.g.
directly be connected with the clinical trial issues. year 2030 knowing very little more than we do now,
While we have already mentioned that very few continuously undermining once-existing standards of
prospective phase III trials are produced (designed care and seriously affecting treatment outcomes of
and executed) by radiation oncologists, another issue thousands of patients.
Clinical Research in Radiation Oncology of Lung Cancer: Why We Fail(ed)? 817

successfully integrate our capabilities and address


6 Possible Solutions burning questions in the field of lung cancer, a
must for current and future generations of radiation
Possible solutions to overcome existing obstacles and oncologists involved in care of patients with lung
improve our ability to undertake meaningful clinical cancer.
research in radiation oncology of lung cancer could
include improvements in education (both residents
and staff radiation oncologists), identification of References
existing weaknesses in research domain which may
present as obstruction to evidence-based oncology
Akerley W, Herndon JE Jr, Lyss AP, Choy H, Turrisi A,
principles, implementation of proven clinical trials Graziano S, Williams T, Zhang C, Vokes EE, Green MR
and meta analyses, especially those favoring radiation (2005) Induction paclitaxel/carboplatin followed by con-
therapy in lung cancer, necessary to produce more current chemoradiation therapy for unresectable stage III
clinical trials by radiation oncologists, especially non-small-cell lung cancer: a limited-access study–CALGB
9534. Clin Lung Cancer 7:47–53
those which combine biology of the disease with the Belani CP, Wang W, Johnson DH (2005a) Phase III study of
technological capabilities of our profession, for which the Eastern Cooperative Oncology Group (ECOG 2597):
(all of the above) we actually need more patients. So, induction chemotherapy followed by either standard
perhaps the very first step on our road towards thoracic radiotherapy or hyperfractionated accelerated
radiotherapy for patients with unresectable stage IIIA and
improvement would be to leave the departmental B non-small-cell lung cancer. J Clin Oncol 23:3760–3767
premises and start both making our achievements Belani CP, Choy H, Bonomi P, Scott C, Travis P, Haluschak J,
more public to both general public and also to other Curran WJ Jr (2005b) Combined chemoradiotherapy regimens
medical professionals. One such successful case has of paclitaxel and carboplatin for locally advanced non-small-
cell lung cancer: a randomized phase II locally advanced
happened in Japan where radiation oncologists multi-modality protocol. J Clin Oncol 23:5883–5891
prominent in stereotactic radiation therapy of early Belderbos J, Uitterhoeve L, van Zandwijk N, Belderbos H,
lung cancer used various means to popularize suc- Rodrigus P, van de Vaart P, Price A, van Walree N, Legrand
cessful efforts to optimize treatments in this disease. C, Dussenne S, Bartelink H, Giaccone G, Koning C (2007)
EORT LCGC and RT Group. Randomised trial of sequen-
That led to more patients refusing surgery and tial versus concurrent chemo-radiotherapy in patients with
requesting stereotactic radiation therapy which, inoperable non-small cell lung cancer (EORTC 08972–
importantly, came with excellent results and further 22973). Eur J Cancer 43:114–121
general popularization of this technique. By making Clamon G, Herndon J, Cooper R (1999) Radiosensitization
with carboplatin for patients with unresectable stage III non-
various (here, meaning both patient and medical small-cell lung cancer: a phase III trial of the Cancer and
professionals!) target groups more aware of our Leukemia group B and the Eastern Cooperative Oncology
capabilities, we may be able to recruit more patients Group. J Clin Oncol 17:4–11
for treatments and, hence, more of those suitable for Curran WJ Jr, Scott C, Langer C, Komaki R, Lee JS, Hauser S
(2000) Phase III comparison of sequential Vs cancer
clinical trials. Such trials should preferably combine (NSCLC): Initial report of Radiation Therapy Oncology
biological and technological aspects of our profession Group concurrent chemoradiation for pts with unresected
in the field of lung cancer. Biological aspects such as stage III non small cell lung (RTOG 9410). Proc Am Soc
irradiation-drug interactions could successfully be Clin Oncol 19:484a (Abstract 1891)
Dillman RO, Seagren SL, Propert KJ, Guerra J, Eaton WL, Perry
explored through translational research using mech- MC (1990) A randomized trial of induction chemotherapy
anistic studies to enlighten the problem of irradiation- plus high-dose radiation versus radiation alone in stage III
drug interactions, but only if translational research is non-small-cell lung cancer. N Engl J Med 323:940–945
understood as ‘‘back and forth’’ from benchmark to El Sharouni SY, Kal HB, Battermann JJ (2003) Accelerated
regrowth of non-small cell lung tumors after induction
bedside and back, aiming to continuously enrich our chemotherapy. Br J Cancer 89:2184–2189
knowledge, performance capabilities and, ultimately, Fried DB, Morris DE, Poole C et al (2004) Systematic review
treatment results. Such clinical studies should ask evaluating the timing of thoracic radiation therapy in
simple questions, ask those questions important for combined modality therapy for limited-stage small-cell
lung cancer. J Clin Oncol 22:4785–4793
radiation oncologists, being biology of the disease- Gandara DR, Chansky K, Albain KS et al (2006) Long-term
driven and being at the same time, technology-adap- survival with concurrent chemoradiation therapy followed
ted. This is the only way we, as a profession, could by consolidation docetaxel in stage IIIB non-small cell lung
818 B. Jeremić

cancer: a phase II Southwest Oncology Group study Le Chevalier T, Arriagada R, Tarayre M, Lacombe-Terrier MJ,
(S9504). Clin Lung Cancer 8:116–121 Laplanche A, Quoix W (1992) Significant effect of adjuvant
Goldstraw P, Crowley J, Chansky K, Giroux DJ, Groome PA, chemotherapy on survival in locally advanced non small
Rami-Porta R, Postmus PE, Rusch V, Sobin L (2007) cell lung carcinoma. J Natl Cancer Inst 84:58 (letter)
International association for the Study of Lung Cancer Movsas B, Moughan J, Komaki R et al (2003) Radiotherapy
International Staging Committee; Participating Institutions. patterns of care study in lung carcinoma. J Clin Oncol
The IASLC Lung Cancer Staging Project: proposals for the 21:4553–4559
revision of the TNM stage groupings in the forthcoming Pijls-Johannesma MCG, de Ruysscher D, Lambin P et al (2004)
(seventh) edition of the TNM Classification of malignant Early versus late chest radiotherapy for limited stage small
tumours. J Thorac Oncol 2:706–714 cell lung cancer. Cochrane Database Syst Rev No. 4
Hanna N, Neubauer M, Yiannoutsos C, McGarry R, Arseneau J, Saunders M, Dische S, Barrett A, Harvey A, Griffiths G, Palmar
Ansari R, Reynolds C, Govindan R, Melnyk A, Fisher W, M (1999) Continuous, hyperfractionated, accelerated radio-
Richards D, Bruetman D, Anderson T, Chowhan N, Nattam S, therapy (CHART) versus conventional radiotherapy in non-
Mantravadi P, Johnson C, Breen T, White A, Einhorn L, small cell lung cancer: mature data from the randomised
Hoosier Oncology Group;US Oncology (2008) Hoosier multicentre trial. CHART Steering committee. Radiother
Oncology Group; US Oncology. Phase III study of cisplatin, Oncol 52:137–148
etoposide, and concurrent chest radiation with or without Sause WT, Scott C, Taylor S, Johnson D, Livingston R, Komaki R
consolidation docetaxel in patients with inoperable stage III (1995) Radiation Therapy Oncology Group 88–08 and Eastern
non small-cell lung cancer: the Hoosier Oncology Group and Cooperative Oncology Group 4588: Preliminary results of a
U.S. Oncology. J Clin Oncol 26:5755–5760 phase III trial in regionally advanced, unresectable nonsmall
Huncharek M, McGarry R (2004) A meta-analysis of the cell lung cancer. J Natl Cancer Inst 87:198–205
timing of chest irradiation in the combined modality Schaake-Koning C, van der Bogaert W, Dalesio O, Festen J,
treatment of limited-stage small cell lung cancer. Oncol- Hoogenhout J, van Houtte P (1992) Effects of concomitant
ogist 9:665–672 cisplatin and radiotherapy on inoperable non-small cell lung
Jeremic B (2006) Timing of concurrent radiotherapy and cancer. N Engl J Med 326:524–530
chemotherapy in limited-disease small-cell lung cancer: Sekine I, Nokihara H, Sumi M, Saijo N, Nishiwaki Y, Ishikura S,
meta-analysis of meta-analyses. Int J Radiat Oncol Biol Mori K, Tsukiyama I, Tamura T (2006) Docetaxel consol-
Phys 64:981–982 idation therapy following cisplatin, vinorelbine, and concur-
Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S (1997) rent thoracic radiotherapy in patients with unresectable stage
Initial versus delayed accelerated hyperfractionated radia- III non-small cell lung cancer. J Thorac Oncol 1:810–815
tion therapy and concurrent chemotherapy in limited small Socinski MA, Blackstock AW, Bogart JA, Wang X, Munley M,
cell lung cancer. J Clin Oncol 15:893–900 Rosenman J, Gu L, Masters GA, Ungaro P, Sleeper A,
Jeremic B, Shibamoto Y, Nikolic N, Milicic B, Milisavljevic S, Green M, Miller AA, Vokes EE (2008) Randomized phase
Dagovic A, Aleksandrovic J, Radosavljevic-Asic G (1999) II trial of induction chemotherapy followed by concurrent
The role of radiation therapy in the combined modality chemotherapy and dose-escalated thoracic conformal radio-
treatment of patients with extensive disease small-cell lung therapy (74 Gy) in stage III non-small-cell lung cancer:
cancer (ED SCLC): a randomized study. J Clin Oncol CALGB 30105. J Clin Oncol 26:2457–2463
17:2092–2099 Turrisi AT 3rd, Kim K, Blum R, Sause WT, Livingston RB,
Langer CJ, Moughan J, Movsas B, Komaki R, Ettinger D, Komaki R, Wagner H, Aisner S, Johnson DH (1999) Twice-
Owen J, Wilson JF (2005) Patterns of care survey (PCS) in daily compared with once-daily thoracic radiotherapy in
lung cancer: how well does current U.S. practice with limited small-cell lung cancer treated concurrently with
chemotherapy in the non-metastatic setting follow the cisplatin and etoposide. N Engl J Med 28(340):265–271
literature. Lung Cancer 48:93–102 Vokes EE, Herndon JE 2nd, Crawford J, Leopold KA, Perry
Lau D, Leigh B, Gandara D, Edelman M, Morgan R, Israel V MC, Miller AA, Green MR (2002) Randomized phase II
(2001) Twice-weekly paclitaxel and weekly carboplatin study of cisplatin with gemcitabine or paclitaxel or vino-
with concurrent thoracic radiation followed by carboplatin/ relbine as induction chemotherapy followed by concomitant
paclitaxel consolidation for stage III non-small-cell lung chemoradiotherapy for stage IIIB non small-cell lung
cancer: a California cancer consortium phase II trial. J Clin cancer: cancer and leukemia group B study 9431. J Clin
Oncol 19:42–447 Oncol 20:4191–4198
Pitfalls in the Design, Analysis,
Presentation, and Interpretation
of Randomized Clinical Trials
Richard Stephens

Contents 11 Has the Result been put into the Context


of the Previous Work in this Area? ...................... 825

1 Introduction.............................................................. 819 12 Conclusions ............................................................... 825

2 Have all the Randomized Patients been References.......................................................................... 825


Accounted for? ......................................................... 820
3 What is the Control Treatment? ........................... 820
Abstract
4 Is the Sample Size based on Information that is Randomised clinical trials are the gold standard
Sensible and Feasible? ............................................ 821 tool for testing new treatments, but need to confirm
5 Has the Trial been Designed to Answer a Clear, to certain basic guidelines in order to be consid-
Unconfounded Question?........................................ 822 ered reliable. Good trial design, analysis, presen-
6 Are the Number of Statistical Tests Limited, tation and interpretation and an awareness of the
and if not, Have the Significance Levels been common pitfalls are likely to maximise the rele-
Adjusted Accordingly?............................................ 822 vance and impact of the trial.
7 Are the Details of the Interim Analyses
and Stopping Rules Clearly Laid Out? ................ 823
8 Have the Hazard Ratio and Especially the 95%
Confidence Interval of the Time-to-Event 1 Introduction
Analyses been given?............................................... 823
9 Are there Predefined Hypotheses for All Key We are in the era of evidence-based medicine, and the
Endpoints? ................................................................ 824
foundation for this evidence is the randomized clini-
10 How has Quality of Life been Assessed cal trial (RCT). In theory RCTs are very simple. Half
and Reported?.......................................................... 825 of the patients receive the standard treatment, the
other half receive the new treatment, and the out-
comes of the two groups are compared. What could
go wrong? Well, in practice, many things, as the
design, conduct, analysis, presentation, and interpre-
tation of RCTs can actually be very complex.
Therefore the importance of high-quality RCTs can-
not be understated. This chapter therefore aims to
alert both those involved with the running of RCTs,
and also those reading papers giving the results of
RCTs, to ten common pitfalls, which may prevent a
R. Stephens (&)
Clinical Trials Unit, London, UK randomized trial from being a true and unbiased
e-mail: richardjamesstephens@gmail.com comparison of the treatments.

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 819
DOI: 10.1007/174_2011_282, Ó Springer-Verlag Berlin Heidelberg 2011
820 R. Stephens

being discovered and more patients deciding to


2 Have all the Randomized Patients withdraw consent. Whilst it can be argued that an
been Accounted for? imbalance of 13:3 can occur by chance, that removing
ineligible patients ensures the trial better reflects the
Given a reasonable number of patients, the beauty of population defined by the protocol, and that including
randomization is that it separates them into perfectly or excluding this number of patients is unlikely to
balanced groups (balanced not only on all their known have a major impact on the results, it is the principle
characteristics, such as age and sex, but also on every that is important, and it raises the question of where to
unknown characteristic as well). Thus it sets up a draw the line: how many patients need to be excluded,
perfect scientific test where the only difference is or what difference in the number of patients excluded
the treatment or management that the patients sub- in each trial arm needs to be seen, before a clinical
sequently receive. Retaining all randomized patients trial can no longer be considered a fair scientific test,
in the trial and in the analysis (the ‘intent-to-treat’ and the results can no longer be considered reliable.
principle), ensures this balance is maintained. Any Whilst some may think it completely illogical to
attempt to improve on this perfect balance between retain any ineligible patients in the analysis, as indi-
the randomized groups is therefore, at best, ill cated above, randomization will ensure that all pos-
advised, and at worst raises concerns about the sible patient characteristics are balanced between the
motives of the researchers. Understanding and groups, including of course, the number of ineligible
applying the intent-to-treat principle is important, as patients, and the number of patients who will poten-
trials need to reflect the policy of choosing to treat a tially decide to drop out of the trial. Therefore, for the
group of patients in a certain way. sake of scientific credibility and complete transpar-
At the time of randomization all patients should ency it is always preferable to retain all randomized
have been considered suitable for the treatments being patients in the trial and report the number (not just the
studied (and thus, once the trial has finished, should proportion) of all patients in all analyses.
reflect the population who are likely to be offered the
treatment). However, papers often list subgroups of
patients who are excluded from the trial and analyses, 3 What is the Control Treatment?
such as those shown to be ineligible by post-
randomization investigations or independent review, In a randomized trial the choice of control treatment
those who do not receive any or all of their protocol is paramount. Logically, it should always be the
treatment, or those not assessed for an endpoint. For current best standard treatment for the condition,
example, Georgoulias et al. (2001) excluded 35 of the although often knowing what is acknowledged as
441 patients randomized and all analyses (which were ‘best’ is difficult. Indeed, there may be situations
claimed to be ‘intent-to-treat’) were then performed where the local, national, and international ‘best’ are
on the remaining 406 patients, and Schiller et al. all different because of, for example, differences in
(2002) excluded 52 patients who were found to be facilities, expertise, or access to drugs. The choice of
ineligible post-randomization in their trial of four control treatment will depend on several factors,
chemotherapy regimens. Removing patients from the including whether the trial result is aimed at affecting
trial or analyses has the potential to upset the perfect local, national, or international practice, how prag-
balance determined by randomization, especially matic the trial is (for example, if the question is ‘does
if there is an imbalance of exclusions across the the addition of radiotherapy to chemotherapy improve
groups. In a recent clinical trial of prophylactic cra- survival?’ the chemotherapy used may not need to be
nial irradiation (PCI) versus observation for patients stated) or how a non-local control treatment will
with locally advanced non-small cell lung cancer affect accrual. It is not difficult to see that the choice
(Gore et al. 2011) 13 PCI and 3 Observation patients of the control treatment can significantly influence the
were excluded from the analysis. Such an imbalance way the trial result is interpreted, as unfortunately
inevitably makes one wonder if those in the PCI much more attention is paid to trials with a ‘positive’
group were seen more often and/or had more detailed result. Thus in order to increase the chances of seeing
investigations that led to more ineligible patients a ‘positive’ outcome, trials can be designed to
Pitfalls in the Design, Analysis, Presentation, and Interpretation 821

compare the new treatment with a poor or inappro- estimation of the true difference. It stands to reason,
priate control. A common trick is to compare the new therefore, that the more patients we study, the better
treatment alone with the new treatment in combina- the estimation. The ‘power’ of a trial relates to the
tion with a standard treatment. Thus in lung cancer chances of identifying a difference if it exists.
there are examples of trials comparing new drug Generally, trials are powered at about 90% (i.e. so that
versus new drug plus cisplatin; for example Splinter a trial has a 90% chance of detecting a difference, if
et al. (1996) compared teniposide with or without one exists), but trials that are underpowered (i.e. do not
cisplatin in advanced NSCLC. Not surprisingly, the include enough patients to reliably detect the differ-
trial showed that the cisplatin combination resulted in ence) are more likely to result in a false-negative result
improved response, progression-free survival and (also referred to as a type II error). Unfortunately, of
overall survival. Such results can be used to claim that course, we never know which ‘negative’ results are
the new combination should be considered an false-negatives! However, all too often sample sizes
effective standard treatment, irrespective of whether are based on what is feasible rather than what is
the new drug actually has any useful effect or not. realistic. For instance, we know that, in lung cancer,
Because of the difficulty, due to the huge number of the addition of a new modality, be it radiotherapy or
patients required, of showing that a new treatment is chemotherapy, to surgery (or supportive care) will
equivalent to a standard treatment, a course of action probably improve survival by only about 5%
sometimes taken is to show that the new treatment is (Non-Small Cell Lung Cancer Collaborative Group
better than a previous standard to the same degree as the 1995). Therefore it is unrealistic to consider that as a
current standard. Thus if new treatment B is 5% better result of tinkering with the drugs, dosages, or sche-
than standard treatment A, the options for another new dule, we are suddenly going to see advantages of a
treatment C are either to try and show that C is equiv- further 10 or 15%. Yet the vast majority of lung cancer
alent to B, or that C is also 5% better than A. However, it trials are based on seeing differences of about 15%,
could be argued that the latter is unethical, as patients which will generally require a sample size of around
are not being offered treatment B, the new current 400 patients. For example, Ranson et al. (2000)
standard of care. Nevertheless, this is a commonly used powered their trial to look for a 100% improvement
strategy. For example, given the NSCLC meta-analysis (from 20% survival at 1 year with supportive care to
(Non-Small Cell Lung Cancer Collaborative Group 40% with paclitaxel), and Sculier et al. (2001), in a
1995) showed a highly significant (P \ 0.0001) sur- three-arm trial considered that a 75% increase might be
vival benefit with cisplatin-based chemotherapy possible with the addition of G-CSF or antibiotics to
over supportive care alone, it could be argued that standard chemotherapy. The rationale for such over-
Roszkowski et al. (2000) should have compared optimistic expectations is that accrual is considered
docetaxel against cisplatin-based chemotherapy rather feasible, whereas aiming for around 1,500 patients to
than supportive care. The fact that they did not, left see a 10% difference or 4,000 patients to see a 5%
unanswered the question of the relative benefits of difference, which is probably the sort of target most
docetaxel compared to cisplatin-based chemotherapy. trials should now be aiming at, is simply considered an
impossible task. Maybe this explains why progress in
lung cancer has been so slow as we have had to wait for
4 Is the Sample Size based meta-analyses to combine data from a number of trials
on Information that is Sensible in order to accumulate the thousands of patients
and Feasible? required to confirm these small differences. A question
then arises as to whether it is ethical to run any trial of
If we had access to every patient with the disease less than perhaps 1,000 patients, given the high prob-
under scrutiny and could randomize them all, we could ability of an inconclusive result. An even greater
obtain a fairly accurate measure of whether the new dilemma occurs with equivalence trials. Taking the
treatment is better than the standard and if so by how same example that the addition of a modality (chemo-
much. However, of course, we do not. We only have therapy) improves survival over surgery alone or sup-
access to a sample of these patients, and all that the portive care by about 5%, what happens when we want
results of our randomized trial can do is to give an to show that a new chemotherapy treatment is as
822 R. Stephens

effective as the standard? If we compare the new che- every 100 ‘positive’ results will be false-positives
motherapy to standard chemotherapy with a trial of 400 (also referred to as a type I error), found purely by
patients the result may suggest no difference but actu- chance. Again, the trouble is we never know which!
ally all we are likely to be able to conclude is that the Whilst we need to be aware that a proportion of
new treatment is somewhere between 15% better and positive trial results may in fact be false-positives
15% worse than the standard, and thus could actually be (and a proportion of negatives false-negatives), the
10% worse than no chemotherapy. Nevertheless, some problem of type I (and type II) errors also affect
papers for example Gatzemeier et al. (2000), claim that analyses within a trial, as the more the tests per-
survival is comparable even though a 20% benefit or formed, the more likely it is that these will be
detriment cannot be ruled out. contaminated with false results. To reduce this risk,
the number of statistical tests performed in a trial
should be limited. A good way of doing this is to
5 Has the Trial been Designed consider that within a trial there is only a certain
to Answer a Clear, Unconfounded amount of P value spending. So, if one test is per-
Question? formed and the result is P B 0.05, then the result can
be considered significant. If two tests are performed
Trials should also always aim to answer only one then perhaps they should only be considered signifi-
clear question. Thus a logical trial design in che- cant if P B 0.025, or as is often used to accommodate
motherapy, where the standard is combination che- interim analyses, the first is only considered signifi-
motherapy would be to add or replace one drug. In cant if P B 0.001 so that the second can be consid-
reality of course such simple designs are not always ered significant if P B 0.049. It is important to be
possible or desirable, as often two widely used aware of how many statistical tests have been per-
treatments need to be compared, or entirely different formed and what is claimed to be significant, as there
schedules or combinations of treatment investigated. can often be a tendency to trawl the data for inter-
Nevertheless, results from trials that change two esting results and significant P values. Consider, for
drugs (or schedules or doses) do not allow one to example, one table relating to the assessment of
tease out the relative value of each changed factor. quality of life (QL) in the paper by Sundstrom et al.
For example, Kelly et al. (2001) compared paclitaxel (2002) where 84 P values were calculated. Statistics
and carboplatin given in 3-weekly cycles with suggest that about four of these will be false-positives
vinorelbine and cisplatin given in 4-weekly cycles. and thus the authors indicated that only P \ 0.01
The trial reported equal efficacy, but if say, a benefit would be considered significant.
had been seen with the paclitaxel/carboplatin sche- Sometimes it is not the number of tests that have
dule would this have been because of the paclitaxel, been performed but the data to which they have been
the carboplatin, or the 3-week cycles? applied that raises concerns. It is, of course, logical
to list the pre-treatment characteristics and to high-
light the balance (or imbalance) between groups.
However, it is illogical to apply a statistical test to
6 Are the Number of Statistical Tests show balance or imbalance in this situation. Statis-
Limited, and if not, Have tical tests are used to estimate the likelihood that an
the Significance Levels been observed difference has not occurred by chance.
Adjusted Accordingly? However, differences in pre-treatment characteristics
can only have occurred by chance, and it is thus an
The P value indicates the probability that an observed inappropriate use of a statistical test and a wasteful
difference has not been found purely by chance. Thus use of P value spending. Recent examples of this
a P value of 0.05 indicates that this result would only unnecessary testing can be found in papers by Tada
have occurred by chance five times in every 100. It is et al. (2004) and Langendijk et al. (2001). If
generally considered that a difference with a P value imbalances in pre-treatment characteristics are
of B 0.05 is a ‘positive’ result. However, it is vital to observed, the analysis of the key endpoints should
remember that this actually means that five out of be adjusted accordingly.
Pitfalls in the Design, Analysis, Presentation, and Interpretation 823

It is important to realize, however, that the P value


reflects statistical significance, and what is important 8 Have the Hazard Ratio
when assessing the results of trials, is clinical sig- and Especially the 95% Confidence
nificance. Given a large enough group of patients, Interval of the Time-to-Event
a tiny difference can be statistically significant, but it Analyses been given?
is the scale of the improvement in outcomes and the
impact on patients that will determine whether clini- Events such as death, progression, and second-line
cians and patients feel a new treatment is worthwhile. treatment should be measured by time-to-event
The commentary by Lee (2011) argues that it is time analyses by constructing Kaplan–Meier curves and
to move away from P values and towards a Bayesian comparing the groups using the log-rank test.
approach. Time-to-event analyses should include all patients
randomized, and be calculated from the date of ran-
domization, since taking the start date as anything
7 Are the Details of the Interim other than randomization (which is the one common
Analyses and Stopping Rules time point for all patients) will have the potential to
Clearly Laid Out? bias the result. For example the date of diagnosis may
not be accurate for all patients, and the date of start of
To ensure patients’ safety it is imperative that the treatment may include different delays for different
accumulating data are reviewed at regular intervals groups, and will not even exist for patients who do not
throughout the trial. Whether ‘regular’ means annu- start treatment.
ally, when accrual reaches certain targets or when a Treatments need to be compared on their overall
certain number of events have occurred, will depend survival because choosing a landmark time point, be
on the trial. This allows decisions to be made it median or 1-year survival, may bias the results. For
regarding closing the trial for reasons such as a sig- instance in a trial of surgery versus a non-surgical
nificant difference being seen, or the likelihood that a intervention, the expectation may well be that the
difference will never be seen. Rules for when the trial surgery group is likely to experience high early post-
should close early must also be agreed and there are a operative mortality but better longer-term survival.
number of options, from fixed P values to Bayesian Thus quoting the survival at say 1 month or 5 years
statements such as ‘the evidence must convince might give an inaccurate picture of the true between-
sceptics’. It is important that among the Data Moni- treatment difference. For example, the shape of the
toring and Ethics Committee (DMEC) members there survival curves seen in the trials reported by Fossella
is knowledge of the disease and treatments and pre- et al. (2000) and Takada et al. (2002) overlap for a
vious DMEC experience, as often DMECs will be considerable time before splitting. Although the
called upon to make very difficult decisions. Clear expected median survival or proportion of patients
indications for the reasons for stopping trials early surviving at key time points is often quoted in pro-
need to be laid out and justified, as there are numerous tocols, these are simply snapshots of the likely sur-
examples where trials have stopped early, but the vivals and the likely survival difference, and are also
results have been unconvincing and new trials have used to calculate a sample size.
had to be set up to clarify the situation. For example, Therefore the hazard ratio (HR) is usually used to
two trials of neo-adjuvant chemotherapy for NSCLC indicate the overall survival difference, although in
(Rosell et al. 1994; Roth et al. 1994) both stopped certain situations, such as when survival curves
early after accruing 60 patients, but subsequently cross, the HR is not appropriate. Conventionally, an
several large trials have been set up to clarify whether HR of \1 indicates that the new treatment is better,
any benefit exists. Few trial reports list stopping rules, and [1 indicates that the new treatment is worse.
but guidelines on stopping suggest that P values of at It is always vital that the 95% confidence interval
least \0.001 need to be seen at interim analyses to (95% CI) around the HR is quoted. This indicates
avoid inappropriate closure. Certainly trials that stop the range in which we are 95% sure that the true
‘because a P value of \0.05 was observed at an value lies. Thus for example in a survival compari-
interim analysis’ need to be questioned. son, an HR of 0.85 with a 95% CI of 0.65–1.05
824 R. Stephens

indicates that our best estimate of the survival dif- deaths without progression, should always be the
ference is that the new treatment is 15% better, but preferred analysis.
we are 95% confident that it is somewhere between Complications also arise when patients have
35% better and 5% worse. This surprisingly wide non-protocol or second-line treatment as effective
range, however, is the sort of range commonly post-protocol treatments can compensate for poor first-
obtained from randomized trials with a sample size line treatments and give a false impression of their
of about 250 patients. Thousands of patients are effectiveness. In such situations, progression-free sur-
required to obtain confidence intervals of only about vival or time to treatment failure analyses need to be
5% around the HR. Even in a trial of more than considered.
1,000 patients, comparing surgery with or without
adjuvant chemotherapy, Scagliotti et al. (2003)
reported an HR of 0.96 with a 95% CI of 0.81–1.31 9 Are there Predefined Hypotheses
indicating that compared to the median survival of for All Key Endpoints?
48 months with surgery alone, adjuvant chemother-
apy could have resulted in a detriment of 5.5 months It is important that all the decisions regarding design
or a benefit of 11 months. issues are clearly stated and justified in the protocol,
One of the difficulties of time-to-event analyses is and also that a detailed analysis plan is written.
what to do with patients who have not experienced the This particularly applies to subgroup analyses,
event. In an analysis of overall survival this is which are only reliable if they are predefined, which
straightforward as patients who are still alive are cen- will usually mean that they are hypothesis driven, and
sored at the time last seen, but analyses looking at a take account of sample size and multiple statistical
specific cause of death, progression, or toxicity have testing. Unless the above rules are respected, subgroup
their own issues. analyses should always be considered with caution and
Although the cause of death may be of interest to treated as only hypothesis generating. All too often,
trialists, to indicate how the treatment is working, when clear overall results are not seen, the data are
survival analyses that only report deaths from cancer trawled for interesting subgroup results and, when
may be interesting but very misleading. For exam- found, hypotheses are built around them. Reporting
ple, a treatment that causes many early treatment- such findings as definitive results is irresponsible.
related deaths may, in a cancer-specific survival It is, of course, often interesting to explore whether
analysis, appear to be the better treatment. Thus both any overall survival difference observed is consistent
Sundstrom et al. (2002) reporting the disease-specific across all subgroups, and analyses stratified for pre-
survival rates in their trial of chemotherapy regimens treatment characteristics are therefore useful; whilst
and Shepherd et al. (2002) in their analysis of pro- Sause et al. (2000) did just that, the subgroup analyses
gression-free survival, censored patients who died did not appear to have been predefined, accounted for
from causes considered unrelated to disease or in the sample size, or considered only as exploratory or
treatment. hypothesis generating. Whilst exploratory analyses are
Many papers purport to show differences between acceptable, analysis by post-randomization factors
treatments in terms of time to progression with the (such as treatment received or response) are totally
use of a Kaplan–Meier plot, taking progression as unacceptable, as the groups being compared may be
the event and censoring those alive (or dead) without defined by the outcome being tested. Thus, for exam-
progression. This sort of analysis can be very mis- ple, comparing the survival of responders versus non-
leading as patients who fail from a competing risk responders is flawed because the responders have to
(for example, an early treatment-related death) that survive long enough to respond. Therefore analyses
precludes the possibility of achieving the event are such as those presented by Fukuoka et al. (2003)
treated the same as censored patients who still have comparing survival by responders, and Socinski et al.
the potential for progression. Examples of this can (2002) showing survival by number of cycles of che-
be found in papers by Sundstrom et al. (2002), motherapy received, must be viewed with great cau-
Ranson et al. (2000) and Pujol et al. (2001). tion. Prognostic factor analyses are sometimes run to
Progression-free survival, which takes into account try and identify the factors most related to survival, but
Pitfalls in the Design, Analysis, Presentation, and Interpretation 825

usually there are far too few patients in a single trial to context of an up-to-date systematic review. Repeating
draw any firm conclusions. For example, in a trial this exercise in 2001, they reported no improvement,
reported by Pujol et al. (2001) multivariate analyses with only three (of 30) trials being so reported
were performed on only 226 patients. In addition, in (Clarke et al. 2002). Such findings are disappointing
most trials, prognostic factor analyses are inappropri- and suggest that there is a general lack of awareness
ate as treatment will be a confounding factor, and that individual trials are only part of the whole pic-
predictive analyses, which look at the impact of treat- ture. We must never lose sight of the fact that lung
ment on subgroups of patients, are needed. cancer is a global problem and without global col-
laboration progress will continue to be painfully slow.

10 How has Quality of Life been


Assessed and Reported? 12 Conclusions

Patient self-assessed QL data are especially difficult All trials and all trial results are important as they
to report reliably because the data are multi- all in some way advance the progress of human
dimensional, longitudinal, and inevitably much is knowledge. Our ultimate aim as trialists is to improve
missing. There are therefore few agreed methods of the treatment of future patients and it is therefore
presenting QL results, and care must be taken to be important that we are as rigorous and honest in our
conservative in making strong claims. Major prob- work as we can be.
lems can arise from starting with inadequate sample
sizes, multiple statistical testing, imputing missing
data, comparing the treatments at time points that References
favor one group, and/or summarizing the data inap-
propriately. Non-standard analyses such as those used Clarke M, Chalmers I (1998) Discussion sections in reports of
by Ranson et al. (2000) estimating separate slopes for controlled trials published in general medical journals:
dropouts and completers, or Sandler et al. (2000) Islands in search of continents? JAMA 280:280–282
Clarke M, Alderson P, Chalmers I (2002) Discussion sections
calculating the change in score from baseline to last in reports of controlled trial published in general medical
observation, should be avoided. journals. JAMA 287:2799–2801
Many of these problems can be mitigated by pre- Fossella FV, DeVore R, Kerr RN, Crawford J, Natale RR et al
defining QL hypotheses which have the effect of (2000) Randomized phase III trial of docetaxel versus
vinorelbine or ifosfamide in patients with advanced non-small
guiding the choice of questionnaire, the choice of cell lung cancer previously treated with platinum-containing
administration time points, the sample size calcula- chemotherapy regimens. J Clin Oncol 18:2354–2362
tion and the analyses to be performed. However, there Fukuoka M, Yano S, Giaccone G, Tamura T, Nagagawa K et al
are few examples of this actually being carried out in (2003) Multi-institutional randomized phase II trial of
gefitinib for previously treated patients with advanced
practice, and consequently, the results from QL non-small cell lung cancer. J Clin Oncol 21:2237–2246
aspects of trials are often disregarded and distrusted Gatzemeier U, von Pawel J, Gottfried M, ten Velde GPM,
by clinicians and patients. Mattson K et al (2000) Phase III comparative study of high
dose cisplatin versus a combination of paclitaxel and
cisplatin in patients with advanced non-small cell lung
cancer. J Clin Oncol 18:3390–3399
11 Has the Result been put Georgoulias V, Papadakis E, Alexopoulos A, Tsiafaki X, Rapti
into the Context of the Previous A et al (2001) Platinum-based and non-platinum-based
Work in this Area? chemotherapy in advanced non-small cell lung cancer: a
randomised multicentre trial. Lancet 357:1478–1484
Gore EM, Bae K, Wong SJ, Sun A, Bonner JA et al (2011)
Trials are rarely islands. Results need to be presented Phase III Comparison of prophylactic cranial irradiation
and discussed in the context of the totality of previous versus observation in patients with locally advanced non-
work. However, Clarke et al. (1998) reviewed the small-cell lung cancer: primary analysis of radiation therapy
oncology group study RTOG 0214. J Clin Oncol 29:
discussion sections of reports of trials published in
272–278
five major journals during one month in 1997 and Kelly K, Crowley J, Bunn PA Jr, Presant CA, Grevstad PK et al
found that only two (of 26) placed their results in the (2001) Randomized phase III trial of paclitaxel plus
826 R. Stephens

carboplatin versus vinorelbine plus cisplatin in the treatment Scagliotti GV, Fossati R, Torri V, Crino L, Giaconne G et al
of patients with advanced non-small cell lung cancer: a (2003) Randomized study of adjuvant chemotherapy for
southwest oncology group trial. J Clin Oncol 19:3210–3218 completely resected stage I, II or IIIa non-small cell lung
Langendijk H, de Jong J, Tjwa M, Muller M, ten Velde G et al cancer. J Natl Cancer Inst 95:1453–1461
(2001) External irradiation versus external irradiation plus Schiller JH, Harrington D, Belani CP, Langer C, Sandler A et al
endobronchial brachytherapy in inoperable non-small cell (2002) Comparison of four chemotherapy regimens for
lung cancer: a prospective randomized study. Rad Oncol advanced non-small cell lung cancer. N Engl J Med
58:257–268 346:92–98
Lee JJ (2011) Demystify statistical significance—time to move Sculier JP, Paesmans M, Lecomte J, van Cutsem O, Lafitte JJ
on from the p value to Bayesian analysis. J Natl Cancer et al (2001) A three-arm phase III randomised trial
103:2–3 assessing, in patients with extensive disease small cell lung
Non-Small Cell Lung Cancer Collaborative Group (1995) cancer, accelerated chemotherapy with support of haema-
Chemotherapy in non-small cell lung cancer: a meta- tological growth factor or oral antibiotics. Br J Cancer
analysis using updated data on individual patients from 52 85:1444–1451
randomised clinical trials. BMJ 311:899–909 Shepherd FA, Giaccone G, Seymour L, Debruyne C, Bezjak A
Pujol J-L, Daures J-P, Riviere A, Quoix E, Westell V et al et al (2002) Prospective, randomized, double-blind, placebo-
(2001) Etoposide plus cisplatin with or without the combi- controlled trial of marimastat after response to first-line
nation of 40 -epidoxorubicin plus cyclophosphamide in chemotherapy in patients with small cell lung cancer: a trial
treatment of extensive small cell lung cancer: a French of the national Cancer Institute of Canada-Clinical Trials
Federation of Cancer Institutes multicenter phase III group and the European Organization for Research and
randomized study. J Natl Cancer Inst 93:300–308 Treatment of Cancer. J Clin Oncol 20:4434–4439
Ranson M, Davidson N, Nicolson M, Falk S, Carmichael J et al Socinski MA, Schell MJ, Peterman A, Bakri K, Yates S et al
(2000) Randomized trial of paclitaxel plus supportive care (2002) Phase III trial comparing a defined duration of
versus supportive care for patients with advanced non-small therapy versus continuous therapy followed by second-line
cell lung cancer. J Natl Cancer Inst 92:1074–1080 therapy in advanced stage IIIb/IV non-small cell lung
Rosell R, Gomez-Condina J, Camps C, Maestre J, Padilla J et al cancer. J Clin Oncol 20:1335–1343
(1994) A randomized trial comparing preoperative chemo- Splinter TA, Sahmoud T, Festen J, van Zandwijk N, Sorenson S
therapy plus surgery with surgery alone in patients with et al (1996) Two schedules of teniposide with or without
non-small cell lung cancer. N Engl J Med 330:153–158 cisplatin in advanced non-small cell lung cancer: a
Roszkowski K, Pluzanska A, Krzakowski M, Smith AP, Saigi E randomized study of the European Organization for
et al (2000) A multicenter, randomized, phase III study of Research and Treatment of Cancer Lung Cancer Cooper-
docetaxel plus best supportive care versus best supportive ative Group. J Clin Oncol 14:127–134
care in chemotherapy-naïve patients with metastatic or non- Sundstrom S, Bremnes RM, Kaasa S, Aasebo U, Hatlevoll R
resectable localized non-small cell lung cancer (NSCLC). et al (2002) Cisplatin and etoposide regimen is superior to
Lung Cancer 27:145–147 cyclophosphamide, epirubicin and vincristine regimen in
Roth JA, Fossella F, Komaki R, Ryan MB, Putnam JB et al small cell lung cancer: results from a randomized phase III
(1994) A randomized trial comparing perioperative chemo- trial with 5 years’ follow-up. J Clin Oncol 20:4665–4672
therapy and surgery with surgery alone in resectable stage Tada H, Tsuchiya R, Ichinose Y, Koike T, Nishizawa N, Nagai
IIIa non-small cell lung cancer. J Natl Cancer Inst 86:673 K, Kato H (2004) A randomized trial comparing adjuvant
Sandler AB, Nemunaitis J, Denham C, von Pawel J, Cormier Y chemotherapy versus surgery alone for completely resected
et al (2000) Phase III trial of gemcitabine plus cisplatin pN2 non-small cell lung cancer (JCOG9304). Lung Cancer
versus cisplatin alone in patients with locally advanced or 43:167–173
metastatic non-small cell lung cancer. J Clin Oncol Takada M, Fukuoka M, Kawahara M, Sugiura T, Yokohama A
18:122–130 et al (2002) Phase III study of concurrent versus sequential
Sause W, Kolesar P, Taylor IV S, Johnson D, Livingston R thoracic radiotherapy in combination with cisplatin and
et al (2000) Final results of phase III trial in regionally etoposide for limited stage small cell lung cancer: results of
advanced unresectable non-small cell lung cancer. Chest the Japan Clinical Oncology Group Study 9104. J Clin
117:358–364 Oncol 20:3054–3060
New Directions in the Evaluation
and Presentation of Clinical Research
in Lung Cancer
Noelle O’Rourke, Fergus Macbeth, and Elinor Thompson

Contents Abstract
In this chapter we summarise the problems in
1 Introduction.............................................................. 827 accessing and summarising the vast body of
2 What the Evidence Tells and clinical research on lung cancer and outline how
Does Not Tell Us ...................................................... 828 research may be made more accessible, efficient
3 Defining the Gaps in Evidence: Formulating and relevant.
Our Questions .......................................................... 829
4 Harnessing the Information to Make It Work
for Us......................................................................... 829
1 Introduction
5 Controlled Clinical Trials as a Route
to Knowledge............................................................ 830
Every lung cancer publication opens with a depress-
6 Systematic Reviews and Meta-Analyses ............... 831 ing list of statistics. Globally lung cancer tops the
7 Cochrane Reviews.................................................... 832 incidence tables—1.61 million cases in 2008 (12.7%
8 Challenge of Novel Technologies ........................... 834 total cancer cases), and also tops mortality with a
consistently high fatality rate across the world—
9 Expanding Our Definition of Trial
Populations ............................................................... 834
1.38 million deaths in 2008, 18.2% of total cancer
mortality (Ferlay et al. 2010). Fifty-five percent of
10 Translating Evidence into Practice
new cases now occur in developing countries, a large
and the Elephant in the Room............................... 835
shift in the past decade, with anticipated continuation
11 What We Need to Do Next..................................... 835 of this trend as rates of cigarette smoking continue to
References.......................................................................... 836 rise in newly industrialised countries. Despite
important developments in surgery, radiotherapy and
chemotherapy over the last 20 years and many hun-
dreds of clinical trials, population-based 5-year sur-
vival rates for lung cancer still range from about 5%
to, at best, 15%. And importantly, all too little is
N. O’Rourke known and published about any changes (for either
Beatson West of Scotland Cancer Centre,
better or worse) in the quality of that survival.
Glasgow, UK
The challenges for lung cancer research in the
F. Macbeth (&)
decade ahead are to improve outcomes not just in
National Institute for Health and Clinical
Excellence (NICE), London, UK terms of added weeks or months of median survival
e-mail: Fergus.Macbeth@nice.org.uk but to increase long-term survival and also to dem-
E. Thompson onstrate improved quality of life by treatment inter-
Univeristy Pompeu Fabra, Barcelona, Spain ventions, especially in the palliative setting where

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 827
DOI: 10.1007/174_2011_299,  Springer-Verlag Berlin Heidelberg 2011
828 N. O’Rourke et al.

survival time is short. The emerging novel technolo- practice that not everyone will agree with even these
gies in radiotherapy delivery for lung cancer present few ‘truths’.
specific research challenges, as these cannot be The following are, we believe, more or less firmly
evaluated easily by the conventional approach of accepted in the management of patients with non-
randomised controlled trials. The increasing lung small-cell lung cancer (NSCLC):
cancer burden of developing nations and indeed the • Radical surgery (lobectomy or pneumonectomy) is
impact of global recession on health-care budgets an effective, potentially curative treatment for
raise further challenges in terms of how to ensure that early-stage disease. Adjuvant chemotherapy fol-
newly introduced drugs and technologies are cost lowing surgery can improve by a few percent the 5-
effective for a population. year survival rates in selected patients.
The massive and growing volume of biomedical • A combination of chemotherapy and radiotherapy
information needs to be harnessed so that we can is effective for patients with unresectable stage III
apply it effectively to our unanswered questions in disease, but there is uncertainty about which drugs
lung cancer management. In this chapter we will and radiotherapy regimens are the best.
reflect on what the evidence from research published • In patients receiving radical dose radiotherapy,
in conventional formats can tell us and what its lim- delivery of chemotherapy concurrent with radiation
itations are. We will then make some suggestions improves survival relative to chemotherapy deliv-
about how we might improve the quality, reliability ered sequentially, though with increased associated
and accessibility of that evidence. We will speculate toxicity.
on the potential offered by evolving information • Cisplatin-based chemotherapy has a modest effect
technologies and by new methodologies in clinical on survival in patients with locally advanced and
research. Finally we will propose the need for struc- metastatic disease but it is uncertain how much
tures to apply the evidence available to clinical overall benefit there is in terms of quality of life.
practice. Second- and third-line chemotherapy in selected
patients may add further small-survival benefit.
And these are the more or less firmly accepted
2 What the Evidence Tells and Does ‘truths’ in the management of patients with small-cell
Not Tell Us lung cancer (SCLC):
• Combination chemotherapy improves survival in
Evidence-based medicine promotes effective use of all patients.
the literature to address areas of clinical uncertainty. • A combination of chemotherapy and thoracic
The critical consumer of the evidence first formulates radiotherapy in those with limited stage disease
a clinical question, then searches the medical litera- improves survival and results in the cure of a small
ture for relevant information, critically analyzes what minority of patients.
is available and finally applies the findings to clinical • Prophylactic cranial irradiation (PCI) for those in
practice. In 1996 David Sackett, one of the early complete remission is likely to be beneficial.
proponents of evidence-based medicine outlined the There is much however that the available evidence
notion of a hierarchy of evidence (Sackett et al. 1996). cannot tell us. Data is accumulating on the effect of
This has since been refined to an established inter- tumour histology and patient genetic profile on the
national standard (Atkins et al. 2004) in which the response to targeted agents. More complete and more
robustness of the evidence is expressed as levels 1–5, precise information is needed on the effects of age,
depending upon the quality of the supporting research sex and co-morbidities on the relative effectiveness
base. This ranges from systematic reviews (level 1) and toxicity of interventions. We also need to
through randomised controlled trials, cohort studies understand better the effects of patient preferences,
and case controls to the variably biased ‘expert perspectives and knowledge on both cure and pallia-
opinion’ (level 5). tion. And given that so few patients with lung cancer
In this way we can list a number of accepted are cured, we must increase our knowledge about
‘truths’ in the treatment of lung cancer. But such is some important and difficult end-of-life issues such as
the contestable nature of clinical science and clinical quality of life during second-line chemotherapy and
New Directions in the Evaluation and Presentation of Clinical Research in Lung Cancer 829

with palliative radiotherapy, when to stop active rt, su, th’ (Drug Therapy, Radiotherapy, Surgery,
treatment and how best to palliate important symp- Therapy)’ gives over 68,000 hits. While MEDLINE is
toms such as breathlessness. a long established and widely used medical database,
its size and breadth may be an obstacle to extracting
relevant information in a filtered and organised
3 Defining the Gaps in Evidence: fashion. Moreover it indexes only about one-third of
Formulating Our Questions all biomedical articles. Key to answering our ques-
tions we need a streamlined information retrieval
The first step in developing the evidence base for lung strategy that will search all possible resources and
cancer care is to formulate those questions to which integrate the diverse datasets obtained. This devel-
we do not currently have clear answers such as: oping field of biomedical informatics has the potential
• Does screening with helical CT scanning reduce to combine the molecular advances in lung cancer
mortality in high-risk populations? research with the ongoing clinical trials data and
• What is the optimal timing and regimen of che- individual patient data. This should enable and
motherapy in patients with Stage-III NSCLC accelerate the ‘bench to bedside’ promise of transla-
undergoing radical dose radiation? tional medicine.
• How much more effective is hyperfractionated, The magnitude and diversity of data generated
hypofractionated and/or accelerated radiotherapy from searching across the whole Life Sciences spec-
than conventional radical radiotherapy? trum necessitates technology which can handle it all.
• What is the role of maintenance chemotherapy in This is becoming a reality with the emergence of
NSCLC? Semantic Web technologies, the latest generation
• Is there a dose–response effect for radical radio- (web 3.0) of web interface applications which permits
therapy in both NSCLC and SCLC? information retrieval based on the properties of the
• What is the optimal management of early-stage data rather than their location within the search
NSCLC in a medically unfit patient: limited surgery environment.
or radical conventional radiotherapy or stereotactic There have been a number of specific initiatives
radiotherapy? launched to promote this translational approach. In
• Do the outcomes from radical radiotherapy the US the NCI has established the Cancer Biomed-
improve with use of new technologies: intensity ical Informatics Grid (CaBIG) which is an open
modulated radiotherapy IMRT, volumetric arc source model network to connect the entire cancer
therapy, particle therapy, image-guided radiother- community for data sharing and with a variety of
apy IGRT? software tools and systems available free to end users
• Will co-registered-PET imaging enable more (www.cabig.nci.nih.gov). In the UK there is the
accurate and effective radiotherapy planning and Oncology Information Exchange (ONIX), a free
improve outcomes? online resource which enables users to search across
And in formulating these questions, we need to diverse datasets from a single point of access.
decide what outcomes we are interested in. Can the Investigators can select the relevant resources for
evidence available answer these questions about sur- their enquiry but tabulate multiple enquiries together
vival but also take account of the patient-oriented and re-use data gathered in this way for combined
outcomes such as patient experience and quality of meta-analyses.
life? Not only has advancing technology offered more
sophisticated means of data searching but the ease
with which this can be accessed and distributed con-
4 Harnessing the Information tinues to progress rapidly. With new hand-held tech-
to Make It Work for Us nologies and internet-enabled mobile phones with
multimedia applications, it is now possible at the
There has been a huge and accelerating growth in bedside to tap into up-to-the-minute current trials,
scientific publications on lung cancer over the last synopses of evidence and clinical guidelines. With so
30 years. Searching Medline for ‘Lung Neoplasms/dt, much information at our fingertips, it is therefore
830 N. O’Rourke et al.

surprising that the number of established ‘facts’ on To achieve a comprehensive and objective analysis
best treatment for lung cancer, as listed above, is of all the controlled trials relating to our clinical
relatively short. It is clear that quantity of data alone question we can undertake a systematic review or
does not produce answers. We need to refine the meta-analysis. These techniques combine data from
information according to its quality and relevance to similar trials so that results (estimates of benefits and
the questions we are asking. harm) from a larger number of people can be evaluated
than have been obtained from individual trials alone.
Even so, a fool-proof search strategy, assessment
5 Controlled Clinical Trials as a Route of trial quality and combined trials analysis do not
to Knowledge provide a lasting guarantee for the evidence base of
our practice. The whole process of data reporting,
Within the hierarchy of clinical evidence, randomised sorting and collating requires continual updating. This
controlled trials rank as the gold standard for indi- is facilitated by the existence of clinical trial registers.
vidual studies. A current search of the Cochrane In 2005 the International Committee of Medical
Central Register of Controlled Clinical Trials identi- Journal Editors (ICJME) issued a joint statement that
fies 6,309 lung cancer trials and the published litera- clinical trials would only be considered for publication
ture runs to many thousands of trial publications. if formally registered from the outset of the study on a
Faced with this vast literature and chasms of clinical trials registry. The WHO facilitated international col-
uncertainty, we need to ensure: laboration on this by defining the minimum dataset for
• an efficient search strategy trial registration. There are now several internationally
• a quality assurance process for what we find recognised registers. Clinicaltrials.gov is sponsored by
• a mechanism for organizing the data in a way that the US National Library of Medicine and currently lists
permits useful analysis. over 100,000 trials across 174 countries with 3,544
An effective strategy to identify trials should lung cancer trials. It does limit trial registration with
search through all the data quickly, finding everything emphasis on federal sponsors or new drug FDA
relevant and only the relevant—that is, both sensitive applications. Current Controlled Trials is a commercial
and specific. Good search strategies will search all the company that also owns the online publishing group
literature, published and unpublished, in all lan- Biomed Central. Each trial on the register of which
guages. They will also focus the search on identifying there are 9,661 at present, has a unique identifier
trials which address specifically the clinical question number (International Standard Randomised Con-
that is being asked. trolled Trial Number register ISRCTN). This system
Decisions as to study quality require the application incorporates a metaRegister (mRCT) which divides the
of criteria for assessing the design. The CONSORT whole set of registered trials into separate registers
(Consolidated Standards of Reporting Trials) group of such as UK National register of cancer trials
scientists and editors established standards for report- (UKCCCR) or NIH trials. Any investigator can access
ing clinical trials in a statement first published in 1996 this registry without subscription, can search for trials
and most recently updated in 2010 with a revised and and can register a new trial. Results of trials can be
elaborated document (www.consort-statement.org). published promptly online by BioMed Central to one
It should make clear in any publication whether the million users. Since 2008 Clinicaltrials.gov has offered
methodology of analysis and reporting is robust, with a the option of reporting summary results online also.
checklist and flow diagram demonstrating adherence to Similarly the Global Trial Bank, a partnership between
these principles. This will enable the reader to confirm the Public Library of Science and the American Med-
that the study was valid and the results were reported in ical Informatics Association, offers a freely accessible
full. It may leave unanswered the question of whether a repository for clinical trial results which has indepen-
small study can be generalised more widely and whe- dent peer-review. (The lack of peer review in some of
ther it can be applied to the individual patient or pop- the sites offering rapid access trial results limits their
ulation that the reader is treating. value).
New Directions in the Evaluation and Presentation of Clinical Research in Lung Cancer 831

A specialised trials register is a database of ran-


domised controlled trials (RCTs) in a particular area 6 Systematic Reviews and
of health care. The aim of a specialised register is to Meta-Analyses
collate the references of all RCTs published, by
means of specially designed, exhaustive searches The pitfalls of traditional ‘expert’ reviews have been
repeated at regular intervals. In addition to searches clearly identified and well described. They are subject
of the world’s major electronic databases to a variety of biases which are usually not explicit and
(e.g., MEDLINE, EMBASE, CINAHL, LILACS, the status of the authors may confer a spurious
Current Contents, Biosis and Index to UK Theses) a authority and validity to the conclusions. These nar-
specialised register also includes references to rative reviews, which may falsely advertise themselves
reports of trials (both on-going and completed) that as ‘systematic’, are characterized by their lack of the
have not been indexed, or that have been published essential components of a systematic review: a focus-
in non-indexed journals, as well as trials found by sed research question, comprehensive search strategy
hand searching of conference proceedings and with explicit methodology, and a rigorous method of
unpublished trials. analysis. The science of systematic reviews, so-called
In its simplest form a specialised register is a ref- ‘secondary’ research (research on the research litera-
erence-based database of citations of RCTs in which ture), and meta-analysis is now well defined with
any one trial may have several references, one for standards in place in regard to their structure and
each article published. A reference-based register is reporting (Preferred Reporting Items for Systematic
the easiest and least resource intensive register to Reviews and Meta-Analyses: www.prisma-statement.
assemble. To construct the register, electronic sear- org). As with clinical trials the PRISMA statement
ches are undertaken using specially developed search advocates registration of all protocols for systematic
strategies, and citations and/or abstracts of all refer- reviews (Booth et al. 2011).
ences identified are then downloaded and reviewed by A meta-analysis refers to the statistical combination
an information specialist to ensure that they meet the (or pooling) of quantitative data from more than one
criteria of the register. A study-based register is more original study, in order to amalgamate results from a
sophisticated, though more resource intensive to larger sample of patients than was available in any of
assemble and update. It uses each individual trial as the individual original studies. A meta-analysis should,
the basic record, rather than every reference pub- therefore have greater statistical power to assess the
lished. If a trial has 100 identified publications, these relative risks and benefits of interventions than the
will be linked to the one record relating to the main individual studies. There are drawbacks associated
trial report. Depending on the level of sophistication with pooling data from different studies, which include
of the register, there may be more detailed data for differences in the populations of patients in the studies,
each study on disease stage, number, age and sex of differences in the interventions given and other dif-
participants, interventions used (including agents in ferences in the studies’ designs. Meta-analyses may be
each arm) and outcome measures assessed. In this undertaken in one of two ways: either by combining the
way an investigator consulting the register would be data as presented in the published reports, or by com-
able to undertake very precise searches—examples of bining the original data on the individual patients
which might include: included in each of the original trials—an individual
• trials of chemotherapy in extensive small-cell lung patient data analysis. Most published meta-analyses
cancer in which cisplatin was included in one arm are reports of pooled data from published reports as
• trials in people over 75 in whom quality of life was these are much quicker and easier to do, although their
measured. findings are less robust.
Importantly a register of lung cancer trials aside An individual patient data meta-analysis (IPD) is a
from its research potential on the data offered, would long and time-consuming process which requires
inform the research strategy on gaps in the evidence contact with the authors of all the original studies to
and target future trial design appropriately. gain access to the original dataset. The data must then
832 N. O’Rourke et al.

be cleaned and re-analysed on all the patients in the


included trials—a process, which often involves many
Collaborative
hundreds if not thousands of patient records. Indi-
Review Centres
vidual patient data analyses are therefore more reli- Groups
able than meta-analyses of published data and these
reviews have been influential in shaping clinical
practice. A recent example in lung cancer is the Steering
Group Methods
comparison of concomitant with sequential radio- Fields
Groups
chemotherapy in locally-advanced NSCLC (Auperin
et al. 2010).
The
A systematic review does not always contain a
Consumer
meta-analysis because the pooling of quantitative data Network
from studies included in the review would not be
feasible—as a result of differences in either the data
itself or the way that data was collected. Systematic Fig. 1 Cochrane collaboration entities
reviews may be either independent/‘ad-hoc’ reviews
or Cochrane Reviews.
subject to peer reviewing by experts in both the rel-
evant clinical field and in the methodology of sys-
tematic reviews. There is a formal mechanism for
7 Cochrane Reviews assessing risk of potential bias in all studies included.
The reviews, all written in a standardised format, each
The Cochrane Collaboration was founded in 1993 to provide recommendations on both research and
promote informed decisions about health care by practice that can be accessed by anyone interested in
dissemination of available scientific evidence. An the topic, be they clinician, health-care consumer,
international, not-for-profit organisation, the Collab- manager, health policy maker or researcher.
oration consists of a network of researchers, health In addition to the topic-focused Cochrane Review
professionals, consumers and others around the world Groups (CRGs) of which the Lung Cancer Review
who work together to prepare, maintain and ensure Group is one, there are also Methods Working Groups
accessibility of systematic reviews of health-care which develop methodology, Fields Groups which
interventions. Working together in collaborative focus on the dimensions of health care such as setting
topic-focused Review Groups, of which there are or type of consumer, the Consumer Network and
currently 53, Cochrane reviewers use a rigorous Cochrane Centres which support people in their
methodology for undertaking extensive searches of geographic location. The whole is co-ordinated by a
published and unpublished research, critically steering group (Fig. 1).
appraising abstracts and articles found, and conduct- Within the Collaboration, all review groups are
ing qualitative and quantitative analyses of the find- expected to develop and maintain their specialised
ings. Cochrane reviews are published in electronic trials register and to download their contents at reg-
format (online and on a CD Rom issued quarterly) as ular intervals into the Cochrane Central Register of
part of the Cochrane Library and versions of reviews Controlled Clinical Trials (CENTRAL). The specia-
may also be published in a peer-reviewed journal lised register is used by Cochrane review groups as a
(Clarke and Horton 2001). The main focus of the fundamental resource to support reviewers in the
Collaboration is on conducting reviews of random- preparation and maintenance of reviews.
ised, controlled trials as the ‘gold-standard’ of sci- The Cochrane Lung Cancer Collaborative
entific investigation (Cochrane 1972). Review Group (LCG) was formed in 1997. The
Cochrane reviews are based on pre-defined rigor- organisational structure includes an international,
ous and explicit methodology as set out in the coordinating editorial team supported by reviewers,
Cochrane handbook. A title is registered followed by translators and consumers. The LCG editorial team
a peer reviewed protocol, and the final article is keeps a list that it has developed in consultation
New Directions in the Evaluation and Presentation of Clinical Research in Lung Cancer 833

Table 1 Cochrane lung cancer review group Table 1 (continued)


Systematic reviews and protocols published in Cochrane 26. Pemetrexed disodium for malignant pleural mesothelioma
Library April 2011 Published protocols
Published reviews Non-small-cell lung cancer
Non-small-cell lung cancer 1. Gemcitabine for NSCLC
1. Post-operative radiotherapy (PORT) for NSCLC 2. Optimal duration of chemotherapy for advanced NSCLC
2. Radical radiotherapy for stage I/II medically inoperable 3. Erlotinib for advanced NSCLC
NSCLC
4. Gefitinib for NSCLC
3. Chemotherapy for NSCLC
5. Taxane schedules for NSCLC
4. Chemo and supportive care versus supportive care alone in
Small-cell lung cancer
advanced NSCLC
6. First-line chemotherapy for synchronous brain mets SCLC
5. Palliative radiotherapy for NSCLC
7. Second-line chemo for SCLC
6. Second-line chemotherapy for NSCLC
General
7. Concurrent chemoradiotherapy in NSCLC
8. Anti-angiogenic therapy for lung cancer
8. Surgery for local and locally-advanced NSCLC
9. Particle therapy versus conventional treatment for lung
9. Prophylactic cranial irradiation for preventing brain
cancer
metastases in patients undergoing radical treatment for
NSCLC New titles
10. Surgery versus radiosurgery for patients with a solitary 1. Pharmacotherapy for the prevention of radiation-induced
brain metastasis pneumonitis
11. Chemotherapy and surgery versus surgery alone in 2. Cisplatin-based versus carboplatin-based chemo in NSCLC
NSCLC 3. Topotecan for advanced NSCLC
12. Second or third additional chemo drug for advanced 4. Topotecan for SCLC
NSCLC
5. PET-CT for assessing mediastinal lymph nodes
13. Palliative endobronchial brachytherapy for NSCLC involvement NSCLC
Small-cell lung cancer
14. Cranial irradiation for preventing brain metastases in
SCLC
with members of the review group in which are
15. Chemotherapy versus best supportive care for extensive
SCLC detailed the titles registered for future systematic
16. Early versus late chest radiotherapy for limited-stage reviews, published review protocols and completed
SCLC reviews. A list of the 26 completed systematic
17. Platinum agents versus non-platinum agents for SCLC reviews and nine published protocols are shown in
General
Table 1. A further five titles have also been regis-
tered (see www.cochrane.es/lcg).
18. Screening for lung cancer
The scope of the LCG covers all aspects of pri-
19. Steroids, radiotherapy, chemotherapy and stents for SVC
mary and secondary prevention, therapy, supportive
obstruction
care, psychological interventions, biological therapy,
20. Surgical sealant for air leak prevention after pulmonary
resection and complementary therapy for lung cancer, other
intra-thoracic tumours (if not addressed by other
21. Drugs for preventing lung cancer
review groups) and metastatic lung disease. Although
22. Pleurodesis for malignant pleural effusion
the remit of the group covers prevention, smoking is
23. Non-invasive interventions for improving well-being and not covered because there is a separate Cochrane
quality of life
Review Group addressing tobacco addiction.
24. Elemene for the treatment of lung cancer
The Cochrane Lung Cancer Group has laid the
Mesothelioma foundation stones and made significant progress in the
25. Radiotherapy for malignant pleural mesothelioma development of an evidence-based, global informa-
(continued) tion resource in lung cancer care.
834 N. O’Rourke et al.

increased efficiency with time with cost per unit


8 Challenge of Novel Technologies activity falling further.
If it is not feasible to conduct a randomised trial,
Systematic reviews, meta-analyses and access to then we must examine other means of appraising the
specialised trials registers offer us the means of new technologies. The most robust method is likely to
evaluating clinical trial data, but the rapidly evolving be a prospective comparative cohort study, which has
new technologies in lung cancer radiotherapy cannot less risk of bias than uncontrolled or retrospective
easily be assessed by these means. In recent years we case series. Indeed this may prove to have better
have had expanding options for radiation treatment external validity than an RCT in that all patients eli-
with IMRT, IGRT, stereotactic radiation, volumetric gible for treatment can be included without the
arc therapy (VMAT), particle beam options with exclusions often required for RCTs. More useful still
protons or heavy ions and a variety of enhanced than a single centre cohort study would be a national
imaging alternatives for radiotherapy planning using or international prospective register of patients treated
4D-CT or functional imaging with PET. These new with this technique, gathering anonymised patient
technologies may have superior biological or physical data. This would also help to define the geographical
characteristics relative to conventional treatment. needs for this technique in terms of capacity and
They may demonstrate improved, more conformal location. With central collection and analysis of data
dose distribution but we cannot make the uncritical across countries there would need to be processes in
assumption that these features guarantee improved place for quality assurance of radiotherapy at con-
outcomes clinically, in the absence of effectiveness tributing centres and for gathering patient demo-
data. graphics and data on cost. The output from this would
There are major theoretical and practical obstacles, be to assess effectiveness and quality issues of the
however to conducting randomised controlled trials in new technique on a large-scale prospective database.
the application of new technology. Society has been
unable to resist the technological imperative over the
years so that if a more sophisticated technology 9 Expanding Our Definition of Trial
comes along with theoretical advantages, then it Populations
should be adopted at the first opportunity. Certainly
the improved dose distribution of new radiotherapy Only a tiny percentage of lung cancer patients
techniques promises the possibility of dose escalation worldwide are entered into randomised controlled
with improved tumour control, without sacrificing trials. Not only does this contribute to the gaps in our
normal tissue complication rates. The small incre- evidence base for clinical practice but it raises the
ments of clinical benefit which might derive from question of external validity of published studies if
such new techniques would not be possible to test in many patients, especially the elderly and those with
an adequately powered clinical trial. Moreover the co-morbidity, are excluded from trials. Can we then
test of how effective the new treatment is might generalise research findings to the patient in front of
depend upon the late toxicity associated with radio- us if our patient is frail and elderly?
therapy or the second malignancy rate, which both We have further difficulty in applying our ‘gold-
require substantially prolonged follow-up. And the standard’ of RCT evidence to the evaluation of new
whole issue of cost of new technology makes evalu- technologies, as described above. There is potential
ation even more problematic, as the major capital and for developing the methodology of systematic
development cost of introducing a new technique reviews to incorporate the types of research that are
would need to precede a clinical trial and so the likely to address new research questions, rather than
cancer centre has to make the commitment in advance limiting our reviews to RCTs. Prospective cohort
of being in a position to judge its effectiveness. Fac- studies may provide the most efficient means of
toring cost into an assessment of effectiveness of new gathering data and measuring outcomes with new
technology is further complicated by continuous treatment techniques. But the largest wealth of
change as cost decreases over time for new technol- clinical data worldwide, as yet untapped, are the
ogies, and the user learning curve means that there is thousands of ‘‘N of 1’’ experiments conducted daily
New Directions in the Evaluation and Presentation of Clinical Research in Lung Cancer 835

by health-care professionals on lung cancer patients domains for assessment. These include whether a
around the world. clear statement is made as to scope and purpose of the
The philosophy of ‘knowledge commons’ has guideline, whether there has been stakeholder
emerged in recent years in the drive to share knowl- involvement, applicability of the guideline to the
edge freely and rapidly on the internet. Cancer population, editorial independence and overall rigor
Commons is an open science initiative for physicians, of development. ‘Consensus’ guidelines derived from
scientists and patients engaged in personalised collected ‘expert’ opinion, like the ad-hoc expert
oncology (www.cancercommons.org). The aim is to reviews, should be objectively assessed for their
individualize treatment based on tumour genomics, to reliability.
then learn as much as possible from each individual The majority of clinical guidelines focus exclu-
patient’s response to treatment and to rapidly dis- sively on the effectiveness of treatment but with rising
seminate these findings to enable other professionals lung cancer incidence in the developing countries and
and patients to use this information in their own ever increasing financial demands in health-care
treatment decisions. The vision is to co-ordinate the worldwide, we can no longer ignore the ‘elephant in
thousands of ‘‘N of 1’’ trials into a giant adaptive the room’. The issue of cost effectiveness of treatment
search for better, individualized cancer treatments. is likely to become a more dominant factor in treat-
ment choices in the years ahead. Funding for new
drugs is already subject to thorough cost-effectiveness
10 Translating Evidence into Practice evaluations by governments and health providers in a
and the Elephant in the Room number of countries. The same scrutiny can be
expected to be applied to all aspects of lung cancer
Our purpose in evaluating clinical research is to guide care. The onus therefore is on researchers to not just
management of individual patients. We strive to offer demonstrate which treatments are most effective but
each patient the best treatment available for their to investigate their cost effectiveness.
individual characteristics and stage of disease, but The UK Department for Health has recently
also to make consistent management decisions across completed a consultation process on value-based
groups of patients. On a national and international pricing for drugs, a new concept in health-care
level this consistency of approach and application of funding which attempts to correlate the cost of the
evidence is achieved by clinical practice guidelines. drug with the actual magnitude of benefit it offers. A
Over recent years a variety of lung cancer guidelines similar proposal is being developed by a worldwide
have been developed by national groups (SIGN non-profit organisation Incentives for Global Health,
(2005) and NICE (2011) in UK, ACCP in US, in its Health impact Fund. This type of cost/benefit
Australian National Health and Medical Research analysis presents particular challenges in lung cancer
Council Guidelines) and by professional associations when the median survival for many patients may be a
(IASLC). small number of months. Some organisations now
As with assessing clinical trials, there are now take account of this in weighting the added value of
formal evaluation systems for judging the methodol- extra months of life when life expectancy is short.
ogy and robustness of recommendations within
treatment guidelines. Those rigorously developed
guidelines will grade the levels of evidence for each 11 What We Need to Do Next
topic 1–5 as described above, with one being highest
level of evidence from systematic reviews. The In conclusion both health-care professionals and
guideline will then propose treatment recommenda- clinical researchers in the field need access to the
tions graded A–D, depending on the level of evidence reliable and up-to-date information on clinical effec-
available to support the recommendation, with grade tiveness that can be provided by systematic reviews
A requiring consistent level 1 evidence to support it. and meta-analyses. A specialised trials register has
The quality of the guideline can be checked using much to offer in accessing trial information. Sys-
systems such as the AGREE criteria (www. tematic reviews of the type promoted and published
agreetrust.org), which identify a number of key by the Cochrane Collaboration should be an essential
836 N. O’Rourke et al.

tool for clinicians, researchers, research funders, gathering and analysing data on all our clinical
health policy makers and anyone with an interest in activity. If we really want to increase our under-
lung cancer prevention, treatment and care. They are standing of the biggest cancer killer of our time, the
tools to improve the quality of clinical practice and means are already at our disposal.
clinical research, to inform a strategic research
agenda and to help streamline a more effective and
efficient use of research funding. But systematic
References
reviews are only as good as the original research from
which they are derived and they can never substitute
Atkins D, Best D et al (2004) Grading quality of evidence and
for poor quality research, absent research or a disor- strength of recommendations. BMJ 328:1490
ganised research direction and agenda. Auperin A, Le Pechoux C et al (2010) Meta-analysis of
One important conclusion which comes from the concomitant versus sequential radiochemotherapy in locally
reviews already published in the Cochrane Library is advanced non-small cell lung cancer. J Clin Oncol
28(13):2181–2190
the need to improve not only the quality of individual Australian National Health and Medical Research Council Guide-
trials (their design and reporting) but also the world- lines. www.nhmrc.gov.au/guidelines/health_guidelines.htm
wide co-ordination of research in lung cancer. Booth A, Clarke M et al (2011) International registry of
Resources are increasingly scarce and the advent systematic review protocols. Lancet 377:108–109
Clarke M, Horton R (2001) Bringing it all together: Lancet-
of new drug treatments as well as evolving radiation Cochrane collaborate on systematic reviews. Lancet
technology will place substantial financial demands 357:1728
on health-care providers in the years ahead. Cochrane AL (1972) Effectiveness and efficiency. Random
The challenge ahead is to answer the important reflections on health services. Nuffield Provincial Hospitals
Trust, London
questions in lung cancer by planning research in a Ferlay J, Shin HR et al (2010) Estimates of worldwide burden
more systematic way. This will enable us to deliver of cancer in 2008: GLOBOCAN 2008. Int J Cancer
individualised optimal treatment for each and every 127:2893–2917
patient. We have the opportunity with innovations in NICE lung cancer guideline (2011). www.nice.org.uk/CG121
Sackett DL, Rosenberg WM et al (1996) Evidence based
information technology to facilitate a much more medicine: what it is and what it isn’t. BMJ 312:71–72
organised, strategic and global approach to wider SIGN lung cancer guideline (2005). www.sign.ac.uk/
recruitment to soundly designed trials and to guidelines/fulltext/80/index.html
Index

(TD0/5), 744 Angiogenic squamous dysplasia (ASD), 47


(TD5/5), 744 Angiopoietin-2, 22
(TD50/5), 744 Angiotensin-converting enzyme (ACE), 235
‘beam’s eye view, 145 Aortopulmonary window, 95
11
C-choline, 182 ARDS, 390
11
C-methionine, 182 Argon plasma coagulation (APC) catheters, 584
18
F-fluoro-deoxyglucose(FDG), 76 Argon plasma coagulation (APC), 50
18
F-fluoro-thymidine (FLT), 76 Arterial oxygen pressure on room air (PO2), 352
201
Tl, 182 Arterioplasty, 109
3D conformal RT (3DCRT), 160 Ataxia, 649
3D planning, 145 Autofluorescence bronchoscopy (AFB), 46
4D computed tomography (4DCT), 148, 347, 728 Avalanche photodiodes (APDs), 78
4-D imaging, 72 Axitinib, 259, 779
4D-cone beam CT (4DCBCT), 160
4D-imaging, 188
a-difluoromethylornithine (DMFO), 654 B
Balloon Dilatation, 584
Basic fibroblast growth factor (b-FGF), 233
A Basic fibroblast growth factor (bFGF/FGF-2), 19
Abraxane, 768 Basic fibroblast growth factor (FGF-2), 654
Accelerated fractionation, 132 Batho and equivalent-tissue-air ratio
Accelerated hyperfractionated radiation therapy, 507 (ETAR) methods, 154
Accelerated hyperfractionation, 498 Bcl-2 Inhibitors, 219, 237, 260
Accelerated repopulation, 144 Bcl-2, 9
Acidic fibroblast growth factor (aFGF/FGF-1), 19 Bevacizumab, 254, 778
Active breathing control (ABC), 148, 347 Bilobectomy, 109
Acute esophagitis, 638 Biological markers, 676, 677
Acute fatigue, 450 Biologically effective dose (BED), 516, 566
Acute myocardial infarction (AMI), 618 Bioreductive drugs, 217
Acute pneumonitis, 611 Blood–brain barrier, 649
Adaptive radiotherapy, 730 Bone marrow, 598
Adenocarcinoma (ADC), 54 Bone metastases, 561
Adenocarcinoma, 446, 812 Bone scans, 71
Adjuvant chemotherapy, 439, 250 Boron neutron capture therapy (BNCT), 649
Age, 446 Bortezomib, 780
AKT Pathway Inhibition, 217 Brachytherapy, 477
ALK targeted inhibitors, 271, 784 Bragg peak, 745, 754
Altered fractionation regimens, 411 Brain metastases, 445, 514, 566, 647
Amifostine (Ethyol), 226 Brain neurotoxicity, 517
Amifostine (WR-2721), 616 Breath-hold techniques, 347
Amifostine, 425, 630, 643 Bronchial artery embolization, 589
Anemia, 600 Bronchial stenosis, 485
Angiogenesis, growth factors, 678 Bronchioloalveolar carcinoma, 55
Angiogenesis, 18 Bronchoplasty, 109

B. Jeremić (ed.), Advances in Radiation Oncology in Lung Cancer, Medical Radiology. Radiation Oncology, 837
DOI: 10.1007/978-3-642-19925-7, Ó Springer-Verlag Berlin Heidelberg 2011
838 Index

B (cont.) CT staging, 446


Bronchoscopic balloon dilatation, 51 CT-based planning, 174
Brown–Sequard syndrome, 628 CTV/PTV, 455
CyberKnife image-guided robotic radiosurgery, 715
Cyclooxygenase COX-2-Inhibitors, 219
C Cyclooxygenase-2 (COX2), 12
Carbogen, 215 Cyclooxygenase-2 Inhibitors, 783
Carbon ion radiation therapy, 754 Cyclopamine, 785
Carbon monoxide (CO), 612 Cytokinesis-block micronucleus (MN) test, 124
Carbon-11 (11C), 76
Carboplatin, 276, 292, 776
Carboplatin/paclitaxel, 249 D
Case controls, 828 Dampening, 347
Cediranib, 259, 779 Data Monitoring and Ethics Committee (DMEC), 823
Celecoxib, 783 Decreased sexual interest, 518
Central airway stenosis (CAO), 46 Deep inspiration breath-hold, 347
Cervical Mediastinoscopy, 94 Dementia, 649
Cetuximab, panitumimab, 775 Demyelination, 629, 650
Cetuximab, 256 Density-correction algorithms, 154
Chart, 411 Depression, 450
Chartwel, 411 Diffusewhite matter changes, 651
Chasing, 347 Diffusing capacity for carbon monoxide (DLCO), 352
Chemical pleurodesis, 586 Diffusion capacity (DLCO), 612
Chemotherapy, 201, 248, 267, 364, 409, 454, 492, Digitally reconstructed radiographs (DRRs), 151
514, 518, 809 Direct machine parameter optimization (DMPO), 702
Chest pain, 454 DNA Repair Inhibitors, 217
Chest radiographs, 64 Docetaxel, 251, 302
Chromogranin A, 801 Dose escalation, 400
Chronic lung fibrosis, 611 Dose intensification, 495
Chronic obstructive pulmonary disease (COPD), 576, 611 Dose-functional histograms (DFHs), 617
Chronic pulmonary dysfunction, 610 Dose-response, 319
Cisplatin, 249, 276 Dose-volume histogram (DVH), 350, 368, 614
Cisplatin/etoposide, 492 Dose-volume relationship, 144
Clinical target volume (CTV), 146, 188, 202, 348, 749, 755 Dynamic and diffusion-weighted
Clinical trials, 524 (DWI) MRI, 72
Clumsiness, 628 Dysphagia, 454, 638
c-MET, 10 Dysphoria, 649
CMR (complete metabolic response), 180 Dyspnea, 575
c-MYC proto-oncogene, 7 Dyspnoea, 450, 454, 551
Cognitive dysfunction, 517
Cognitive functioning, 518
Cohort studies, 828 E
Colony formation method, 120 Early Stage Non-Small-Cell Lung Cancer, 525
Combined modality treatment, 434 Echinoderm microtubule-associated protein-like 4
Computed tomography (CT), 64, 90, 173, 202 (EML4)-anaplastic lymphoma kinase (ALK)
Computer aided detection (CAD), 67 fusion oncogene (EML4-ALK), 6
Concurrent Radiochemotherapy, 415 Echinoderm Microtubule-Associated Protein-Like 4-Anaplastic
Cone beam CT (CBCT) , 736, 346 Lymphom Kinase Translocation, 800
Consolidation chemotherapy, 420, 810 Edema, 650
CONSORT (Consolidated Standards of ReportingTrials), 830 Efaproxaril, 237
Continuous hyperfractionated accelerated radiation therapy Efaproxiral (RSR-13), 215
(CHART), 132 Elderly, 524, 812
Control treatment, 820 Elective nodal irradiation, 203
Conventional radiation therapy, 316 Elective nodal irradiation (ENI), 192
Convolution-superposition (CS) method, 154 Electrocautery (EC), 49
Cough, 454, 518, 551, 575 Electromagnetic navigation (EMN) bronchoscopy, 46, 48
COX-2 inhibitors, 231, 271 Electronic portal imaging device (EPID), 694
Crizotinib, 259, 784 Electronic portal imaging devices (EPIDs), 738
Cryotherapy, 52, 483, 583 Emotional functioning, 518
Cryptogenic organizing pneumonia (COP), 122 En bloc chest wall resection, 109
CT on rails, 346 Endobronchial brachytherapy (EBBT), 52, 556
Index 839

Endobronchial Prosthesis, 580 Granulocyte colony stimulating factor, 19


Endobronchial ultrasonography (EBUS), 71 Granulocyte-colony stimulating factors (G-CSFs), 603
Endobronchial ultrasound (EBUS), 46, 94 Gross tumor volume (GTV), 145, 188, 202, 348, 455,
Endoluminal brachytherapy, 477 748, 755, 813
Endoscopic (esophageal) ultrasound (EUS), 90 GyE (Gray equivalents), 754
Endoscopic ultrasound, 71
Enhancement of tumour response, 268
Epidermal growth factor recepto proto-oncogenes: H
c-erb-B1, 796 Hazard ratio (HR), 823
Epidermal growth factor receptor (EGFR), 22, 57, 234, 775 HDAC inhibitors, 271, 781
Epidermal growth factor receptor (EGFR; ErbB-1), 11 Heavy particle beams, 754
Equivalent uniform dose (EUD), 699 Heavy-charged particle, 746
Equivalent-pathlength (EPL) model, 154 Hedgehog inhibitors, 260
Erlotinib, 252, 775, 777 Hedgehog Pathway Inhibitors, 260, 271, 785
Erythropoiesis-stimulating agent (ESA), 605 Hematopoietic growth factors, 589
Erythropoietin (EPO), 216 Hematopoietic stem cells, 589
Esophageal Endoscopic Ultrasound (EUS), 94 Hemiparesis, 628
Esophageal ultrasonography (EUS), 71 Hemoglobin (Hb), 604
Esophagitis index, 639 Hemoptysis, 454, 485, 551, 575
Etoposid, 260, 276, 773 Hemorrhage, 390
Everolimus, 782 Hepatocyte growth factor, 22
Evidence-based medicine, 819, 828 Hepatocyte growth factor/scatter factor (HGF/SF), 19
Extended cervical mediastinoscopy, 95 High linear energy transfer (LET), 754
Extensive disease small cell lung cancer, 505, 537, 813 High-dose chemotherapy, 494
Extremity weakness, 628 High-dose rate brachytherapy, 477
High-dose rate endobronchial brachytherapy (HDR-EBRT), 52
High-molecular-weight mucin-like antigen KL-6, 120
F Histology, 812
FACT-L, 664 Histone deacetylase inhibitors, 781
FACT-TOI, 664 Histone deacetylases (HDAC), 218
FDG-PET, 189 Horners syndrome, 454
Febrile neutropenia (FN), 599 Human manganese superoxide dismutase transgene, 644
Fibroblast growth factor-3 (FGF-3/ int-2), 19 Human MutS Homologue and Human MutL
Fibroblast growth factor-4 (FGF-4/hst/K-FGF), 19 Homologue 1, 800
Fiducial markers, 344, 739 Human recombinant keratinocyte growth factor, 644
Figitumumab, 782 Hybrid Stents, 582
Fine motor coordination, 517 Hyperbaric oxygen (HBO), 633, 655
Flavopiridol (alvocidib), 236 Hyperbaric Oxygen, 214
Flexible intraoperative template (FIT), 472 Hyperfractionation, 132, 498
FLT, 182 Hypoxia, 184, 214, 576
Fluorescence, 374 Hypoxic Cell Radiosensitizers, 216
Forced expiratory volume in one second (FEV1), 352, 612
Forced vital capacity (FVC), 352, 612
Four dimensional computed tomography (4D-CT), 692 I
Four-dimensional (4D)-CT scanning approach, 196 Idiopathic interstitial pneumonitis, 120
Four-dimensional radiotherapy (4DRT), 157 Ifosfamide, 277
Frontal lobe dysfunction, 517 IGF-1R pathway inhibitors, 260
Functional Assessment of Cancer Therapy-General IGF-IR targeting therapies, 271
(FACT-G), 664 Interleukin [IL]-1b, 231
Interleukin 1 (IL-1), 120, 613, 629, 649
IL-6, 615
G Interleukin-8, 19
Gamma linolenic [omega-6] acid, 653 Image guided radiotherapy (IGRT), 725, 736
Gating, 347 Imaging, 148
Gefitinib, 255, 777 Increased vascular permeability, 650
Gemcitabine, 251, 297 Independent cell kill, 268
Gender, Age and Ethnicity, 679 Individual patient data meta-analysis (IPD), 831
Gene expression profiles, 796 Induction chemotherapy, 413, 414, 439, 440, 810
Genetic tumor factors, 677 Inhibitors to poly(ADP-ribose) polymerase (PARP), 237
Geographic miss, 144 Insulin-like growth factor (IGF), 630
Geriatric assessment, 524 Insulinlike growth factor-1 (IGF-1), 654
840 Index

I (cont.) Location, 812


Intellectual decline, 649 Locoregional post-surgical recurrences, 545
Intensity modulated (IMRT), 367 Locoregional recurrence of small-cell lung cancer, 556
Intensity-modulated radiation therapy (IMRT), 133, 401, 424, Low-dose CT screening, 65
528, 692 Low-dose daily chemotherapy, 507
Intensity-modulated arc therapy (IMAT), 692 Lung Cancer Symptom Scale (LCSS), 664
Intensity-modulated proton therapy, 747 Lung metastases, 568
Intercellular adhesion molecule-1 (ICAM-1), 649 Lung tumors, 119
Internal margin (IM), 755 Lymph node dissection, 92
Internal target volume (ITV), 188, 203
International Commission on Radiation Units
(ICRU), 188, 202 M
International Commission on Radiation Units Magnetic resonance imaging(MRI), 90
and Measurements (ICRU), 145 Malacia, 629
Inter-observer variability, 188 Manganese superoxide dismutase (MnSOD), 238
Interstitial brachytherapy, 477 Markers of cellular adhesion, 677
Interventional pulmonology, 45 Markers of DNA Repair, 798
Intracranial stereotactic radiosurgery (SRS), 716 Markers of tumor proliferation, 677
Intraoperative brachytherapy, 470 Matrix metalloproteinases, 22
Intraoperative interstitial brachytherapy, 477 Maximal intensity projection (MIP) approach, 702
Intraoperative nodal staging, 92 Maximum intensity projection (MIP), 165, 197
Intraoperative radiation (IORT), 462 Maximum tolerated dose (MTD), 402
Intrapulmonary recurrence, 545 Mean lung dose, 121
Intratumoral microvessel density (MVD), 20 Mediastinal lymph node recurrences, 545
Invasive mediastinal staging, 92 Mediastinal lymph node sampling, 92
Irinotecan, 251, 303 Mediastinotomy, 95
Irradiation volume, 499 Medical Outcomes Study (MOS), 663
Irritability, 518 Medical Outcomes Study 36 item Short Form Health Survey
ITV, 348 (MOS SF-36), 664
Megavoltage EPIDs, 738
Memory loss, 649
K Meta-Analyses, 515, 831
Keratinocyte growth factor (KGF), 233 Metallic Stents, 581
Kilovoltage EPIDs, 738 Metastasis, 516
Metastatic Non-Small-Cell Lung Cancer, 532
Metastatic spinal cord compression, 561
L Misonidazole, 216
Lack of energy, 518 Mitogen-activated protein (MAP) kinase, 218
Large cell carcinoma, 54, 446, 812 Mitomycin, 277
Late esophageal damage, 639 Mobile linear accelerator, 464
Late toxicity, 611 Molecular markers, 794
Lethargy, 649 MRI (Magnetic Resonance Imaging), 64
Leukopenia, 599 M-Staging, 81
Lhermitte sign, 628 mTOR inhibitors, 260, 271, 782
Light Emitting Diodes (LED), 373 Multileaf collimator (MLC), 149
Light initiation interval (DLI), 374 Multi-leaf collimators (MLCs), 692
Light source, 372 MVCT, 728
Limited disease small-cell lung cancer, 533, 492, 815 Myelosuppression, 589
Limited stage disease, 201
Linear analogue self-assessment [LASA] scales, 667
Linear-quadratic (LQ) formula, 350 N
Line-of-response (LOR), 76 Nanotechnology, 272
Linifanib, 259 Narrow band imaging bronchoscopy, 46
Liver metastases, 568 Nd: YAG laser, 483
Lobectomy, 109, 249, 367, 439 Nd:YAG Laser Photoresection, 48
Local control, 144 Neo-adjuvant chemotherapy, 250, 447
Locally advanced non-small cell lung cancer Neurocognitive Function, 450
(LA-NSCLC), 445 Neuron-Specific Enolase, 801
Locally Advanced Nonsmall-Cell Lung Cancer, 409, 529 Nimorazole, 216
Locally recurrent lung cancer, 546 Nitrogen-13 (13N), 76
Locally recurrent non-small-cell lung cancer, 544 Non small-cell lung cancer (NSCLC), 54, 515
Index 841

Non-coplanar beams, 344 Potential doubling time (Tpot), 124


Non-small cell carcinoma (NSCLC), 54, 524, 812 Pre-chemotherapy volume, 190, 499
Normal lung tissue, 119 Pro-Gastrin-Releasing Peptide, 801
Normal tissue complication probability (NTCP), 144, 403 Prognostic factors, 327, 676
N-stage, 81, 90 Proliferative activity, 124
Numbness, 628 Prophylactic cranial irradiation (PCI), 446, 492,
506, 514, 647
Prostaglandin E2 (PGE2), 231
O Protease inhibitors, 271
Odynophagia, 638 Proteasome Inhibitors, 780
Once-daily thoracic radiation, 498 Protection of normal tissues, 268
Organs at risk (OARs), 692 Proton-beam dosimetry, 746
Overall treatment time, 498 Protons, 744
Oxygen, 214 Proto-oncogene KRAS, 8
Oxygen-enhancement ratio (OER), 214 Pulmonary bronchitis, 485
Pulmonary function tests(PFTs), 612

P
p53 Tumor Suppressor Gene, 795 Q
p53, 12 QLQ-LC13, 664
Paclitaxel, 292 Quality of Life Questionnaire Core 30 (QLQ-C30), 664
Palifermin, 644 Quality of life (QOL), 661, 332, 450
Palliative radiation therapy, 533 Quality-adjusted life expectancy (QALE), 518
Palliative radiotherapy, 454 Quality-adjusted survival (QAS), 295
Paraneoplastic syndromes, 492
Paresthesias, 628
PARP inhibitors, 271, 780, 218 R
Pathological fractures, 561 Radiation induced brain toxicity, 648
Patient immobilization, 344 Radiation myelopathy, 627
Patient-reported outcomes (PROs), 662 Radiation pneumonitis, 121, 754
Patient-Reported Outcomes Measurement Information System Radiation response, 119
(PROMIS), 670 Radiation Sensitizers, 213
Pemetrexed, 251, 304, 766 Radiation therapy, 267, 249, 410, 492, 525, 809
Pentoxifylline (Trental), 234 Radiation-induced liver disease (RILD), 569
Performance Status, 680 Radiation-induced lung toxicity, 609
Perfusion and ventilation scans, 612 Radiochemotherapy, 410, 529
Perioperative high-dose-rate brachytherapy (PHDRB), 471 Radiofrequency ablation, 382
Permanent interstitial transbronchial implantation, 478 Radiopharmaceuticals, 271, 785
Personalized medicine, 794 Radioprotectors, 424
PET (Positron Emission Tomography), 64 Radiosensitivity, 124
PET- CT, 64 Radiosensitizer, 214
PET/MR imaging, 77 Radiosurgery, 568
Photodynamic reaction (PDR), 372 Radiotherapy planning, 177
Photodynamic Therapy (PDT), 52, 371, 483, 583 Radiotherapy, 439, 454, 514, 518
Photomultipliers (PMTs), 77 Radivnecrosis, 651
Photosensitizing agent (PS), 372 Randomised controlled trials, 828
Planning target volume (PTV), 146, 188, 203, 348, Randomized clinical trial (RCT), 819
748, 755 Ras Oncogenes p21, 794
Platelet-derived growth factor (PDGF), 19, 120, 654 Recombinant human erythropoietin
Pleuropneumonectomy, 109 (r-HuEPO epoietin-alfa), 604
PMD (progressive metabolic disease), 180 Recombinant interleukin-11 (rIL-11), 234
PMR (partial metabolic response), 180 Recurrence, 544
Pneumonectomy, 109, 249, 367, 390, 439 Reirradiation of Small-Cell
Polo-like kinase inhibitors, 271, 784 Lung Cancer, 556
Poly(ADP-Ribose) Polymerase Inhibitors, 236 Reirradiation, 549
Porphyrins, 372 Relative mean lung dose (rMLD), 404
Positron emission tomography (PET), 75, 92, 147, Reoperation, 545
173, 202 Residual volume(RV), 612
Post-chemotherapy volumes, 190, 499 Respiratory correlated PET-CT, 347
Postoperative radiation therapy (PORT), 364 Respiratory gated therapy, 148
Postoperative Radiotherapy, 194 Respiratory Gating, 183
842 Index

R (cont.) TGF-b, 613, 615


rh-MnSOD (manganese superoxide dismutase), 654 Therapeutic ratio, 268
Ribonucleotide-Diphosphate Thoracentesis, 585
Reductase M1, 799 Thoracic radiation therapy, 506
Thoracic radiotherapy, 201, 364
Thoracoscopy, 585
S Thoracotomy, 90
Search strategy, 830 Three-dimensional conformal radiation therapy, 188, 317, 403,
Segmentectomy, 109 424, 455, 692
Sequential radiochemotherapy, 412 Thrombocytopenia, 599
Serine/threonine kinase 11 (STK 11), 7 Thrombopoietin (TPO), 605
Serological Tumor Markers, 678 Thymidine phosphorylase, 22
Serum Proteomic Profiling, 801 Thyroid transcription factor 1 (TTF-1), 5
Set-up error, 197 Time-to-event analyses, 823
Set-up uncertainty, 146 Tirapazamine, 217
Shortness of breath, 518, 610 Tiredness, 518
Sickness Impact Profile (SIP), 663, 664 TNF-a PDGF, 613
Silastic Stents, 580 TNFa, 629
Simultaneous integrated boost(SIB), 699 Tomotherapy, 725
Single nucleotide polymorphism (SNP) arrays, 4 Topotecan, 260
Single-photon-emission computed Total lung capacity (TLC), 612
tomography(SPECT), 612 Toxicity, 328, 450, 532
Sleeve resection, 109 Tracheobronchial stents, 51
Slow CT scanning, 196 Tracheoesophageal Fistula, 587
Slow CT, 347 Transbronchial Needle Biopsy (TBNBx), 93
Small molecule drugs, 32 Transesophageal ultrasound, 64
Small-cell carcinoma (SCLC), 54, 201, 492, 514, 533 Transforming growth factor-a(TGF-a), 19
SMD (stable metabolic disease), 180 Transforming growth factor-b(TGF-b), 19
Sorafenib, 259, 779 Translational research, 794
Spatial cooperation, 268 Transthoracic Needle Biopsy (TTNBx), 93
SPECT, 182 Treated volume, 455
Squamous carcinoma, 446 Treatment planning, 144
Squamous cell carcinomas, 812 Treatment Volume, 323
Stage, 454, 812 Trial outcome index, 664
Standard radiation therapy, 411 Tri-modality approach, 436
Standardized uptake (SUV), 180, 190 T-stage, 90
Stereotactic ablative radiotherapy (SABR), 715, 344 T-staging, 80, 90
Stereotactic body radiation therapy (SBRT), 344, 569, 692, Tumor angiogenesis, 19
715, 728 Tumor histology, 677
Stereotactic fractionated radiation therapy, 424 Tumor motion, 148
Stereotactic radiation therapy, 528 Tumor necrosis factor a (TNF-a), 19, 231, 649
Stereotactic radiosurgery (SRS), 647 Tumor size, 322
Subjective significance questionnaire (SSQ), 666 Tumor stage, 322
Sucralfate, 644 Tumorcontrol probability, 144
Sunitinib, 259, 779 Tumour size, 454
Superior sulcus tumors, 434 Twice-daily tho racic radiation, 498
Superior vena cava syndrome, 454 Two-dimensional radiotherapy (2DRT), 454
Surgery, 315, 364, 545, 809 Type I error, 821
Surgical resection, 647 Type II error, 822
Surgical staging, 89 Type II pneumocyte, 120
Symptomatic relief, 549 Tyrosine kinase inhibitors (TKIs), 775
Syncope, 450
Systematic error, 188
Systematic reviews, 828, 831 V
V/Q (Ventilation and Perfusion)
Scans, 71
T V20, 121
Target volume, 202 V20, 350
Targeted gene therapy, 32 V5, 121
Temozolamide, 780 Vandetanib, 259, 779
Temporary implants, 480 Vascular disrupting agents, 32
Index 843

Vascular endothelial growth factor (VEGF), 18, 257, 629 W


Vatalanib, 259 Weakness, 450
Verbal memory, 517 Wedge resection, 109
Video-assisted thoracic surgery Weight loss, 454, 680
(VATS), 90, 95, 112, 249 White matter necrosis, 629
Vinblastin, 277 Whole-brain radiotherapy (WBRT), 566, 647
Vinorelbine, 249, 297
Vital capacity (VC), 612
Volumetric-modulated arc therapy (VMAT), 692 Z
Vorinostat, 781 Zileuton, 783

You might also like